Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Minerva Endocrinol ; 31(1): 13-26, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16498361

ABSTRACT

Estrogens are biosynthesized from androgens by the cytochrome P450 enzyme complex called aromatase. Aromatase is found in several tissues in the body and aromatase (CYP19) gene expression is regulated in a tissue-specific manner via use of alternative promoters. Aromatase transcript expression and enzyme activity in breast tumor tissue is greater than that in the normal breast tissue, and prostaglandins can increase CYP19 expression and aromatase activity in breast cancer cells. Cyclooxygenase (COX) is a key enzyme in the production of prostaglandins. Higher levels of COX-2 isoform were observed in breast cancer tissue when compared to normal breast tissue, and a strong association between CYP19 gene expression and the expression of COX genes are found. Epidemiological studies have shown that nonsteroidal anti-inflammatory drugs (NSAIDs) may be beneficial in reducing the incidence of breast cancer, although the exact mechanisms remain unclear. Both NSAIDs and COX-selective inhibitors suppress aromatase mRNA expression and enzyme activity in breast cancer cells. Real time PCR results demonstrate that this suppression occurs through regulation of CYP19 expression involving promoters I.4 and II, the promoters involved in the development of breast cancer. High levels of COX-2 expression result in higher levels of prostaglandin E2 (PGE2), which in turn increases CYP19 expression through increases in intracellular cyclic AMP levels and activation of promoter II. Thus, PGE2 produced by COX enzymes may act locally in paracrine and autocrine fashion to increase the biosynthesis of estrogen by aromatase in hormone-dependent breast cancer development.


Subject(s)
Aromatase Inhibitors/therapeutic use , Aromatase/physiology , Breast Neoplasms/enzymology , Cyclooxygenase Inhibitors/therapeutic use , Estrogens/physiology , Neoplasms, Hormone-Dependent/enzymology , Prostaglandin-Endoperoxide Synthases/physiology , Adipose Tissue/enzymology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arachidonic Acids/metabolism , Aromatase/genetics , Aromatase Inhibitors/pharmacology , Breast Neoplasms/prevention & control , Cell Transformation, Neoplastic , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/physiology , Female , Gene Expression Regulation, Enzymologic/drug effects , Growth Substances/physiology , Humans , Male , Neoplasms, Hormone-Dependent/prevention & control , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/genetics , Regulatory Sequences, Nucleic Acid
2.
Am J Ther ; 8(5): 333-44, 2001.
Article in English | MEDLINE | ID: mdl-11550075

ABSTRACT

Estrogens are involved in numerous physiologic processes and have crucial roles in particular disease states, such as mammary carcinomas. Estradiol, the most potent endogenous estrogen, is biosynthesized from androgens by the cytochrome P-450 enzyme complex called aromatase. Aromatase is found in breast tissue, and the importance of intratumoral aromatase and local estrogen production is being unraveled. Inhibition of aromatase is an important approach for reducing growth stimulatory effects of estrogens in hormone-dependent breast cancer. Effective aromatase inhibitors have been developed as therapeutic agents for controlling estrogen-dependent breast cancer. Investigations into the development of aromatase inhibitors began in the 1970s and have expanded greatly in the past three decades. Competitive aromatase inhibitors are molecules that compete with the substrate androstenedione for noncovalent binding to the active site of the enzyme to decrease the amount of product formed. Steroidal inhibitors that have been developed to date build on the basic androstenedione nucleus and incorporate chemical substituents at varying positions on the steroid. The structure-activity relationships for steroidal inhibitors have become more refined in the past decade, and only some modifications can be made to the steroid and still keep its affinity for aromatase. Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Mechanism-based aromatase inhibitors are inhibitors that mimic the substrate, are converted by the enzyme to a reactive intermediate, and result in the inactivation of aromatase. Aromatase inhibitors, both steroidal and nonsteroidal, have shown clinical efficacy for the treatment of breast cancer. The initial nonselective nature of nonsteroidal inhibitors such as aminoglutethimide has been greatly reduced in the later generations of inhibitors, anastrozole and letrozole. Mechanism-based steroidal inhibitors such as 4-hydroxyandrostenedione and exemestane produce prolonged aromatase inhibition in patients. The potent and selective third-generation aromatase inhibitors anastrozole, letrozole, and exemestane are approved for clinical use as second-line endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies.


Subject(s)
Aromatase , Breast Neoplasms , Enzyme Inhibitors/therapeutic use , Aromatase/metabolism , Aromatase/physiology , Aromatase Inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Female , Humans
3.
J Steroid Biochem Mol Biol ; 78(2): 145-56, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11566439

ABSTRACT

An estradiol metabolite, 2-methoxyestradiol (2-MeOE(2)), has shown antiproliferative effects in both hormone-dependent and hormone-independent breast cancer cells. Previously, a series of 2-hydroxyalkyl estradiol analogs had been synthesized in our laboratories as potential probes for comparison of estrogen receptor (ER)-mediated versus non-ER-mediated effects in breast cancer cells. A methoxy derivative of 2-hydroxymethyl estradiol was prepared for biological evaluation and comparison with 2-MeOE(2). Estrogenic activity of the synthetic analogs was evaluated in two ways, one by examining affinity of the analogs for the estrogen receptor in MCF-7 cells and the other by examining the ability of the analogs to induce estrogen-responsive gene expression. The analog, 2-methoxymethyl estradiol (2-MeOMeE(2)), demonstrated weak affinity for the estrogen receptor (0.9% of estradiol) and weak ability to stimulate estrogen-induced expression of the pS2 gene (0.02% of estradiol). Antitumor activity was evaluated both in vitro and in vivo. The steroidal nucleus seems to be an attractive target for developing novel tubulin polymerization inhibitors. Additionally, such steroidal compounds may have low toxicity compared to the natural products known to interact with tubulin. Interestingly, 2-MeOMeE(2) inhibited tubulin polymerization in vitro at concentrations of 1 and 3 microM and was more effective than 2-MeOE(2). In cells, 2-MeOMeE(2) was effective in suppressing growth and inducing cytotoxicity in MCF-7 and MDA-MB-231 breast cancer cells. The cytotoxic effects of 2-MeOMeE(2) are associated with alterations in tubulin dynamics, with the frequent appearance of misaligned chromosomes, a significant mitotic delay, and the formation of multinucleated cells. In comparison, 2-MeOE(2) was more effective than 2-MeOMeE(2) in producing cytotoxicity and altering tubulin dynamics in intact cells. Assessment of in vivo antitumor activity was performed in athymic mice containing human breast tumor xenografts. Nude mice bearing MDA-MB-435 tumor xenografts were treated i.p. with 50 mg/kg per day of 2-MeOMeE(2) or vehicle control for 45 days. Treatment with 2-MeOMeE(2) resulted in an approximate 50% reduction in mean tumor volume at treatment day 45 when compared to control animals and had no effect on animal weight. Thus, 2-MeOMeE(2) is an estrogen analog with minimal estrogenic properties that demonstrates antiproliferative effects both in vitro and in the human xenograft animal model of human breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Estradiol/pharmacology , Estrogens/pharmacology , Proteins , 2-Methoxyestradiol , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents, Hormonal/chemical synthesis , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/chemical synthesis , Estrogens/chemical synthesis , Female , Humans , Mice , Mice, Nude , Protein Biosynthesis , Receptors, Estrogen/metabolism , Trefoil Factor-1 , Tubulin/drug effects , Tumor Cells, Cultured , Tumor Suppressor Proteins
4.
Gene Expr ; 9(4-5): 157-71, 2001.
Article in English | MEDLINE | ID: mdl-11444526

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a potent inhibitor of growth and proliferation of breast epithelial cells, and loss of sensitivity to its effects has been associated with malignant transformation and tumorigenesis. The biological effects of TGF-beta are mediated by the TGF-beta receptor complex, a multimer composed of TGF-beta receptor type I (TbetaR-I) and TGF-beta receptor type II (TbetaR-II) subunits. Evidence suggests that loss of expression of Tbeta3R-II is implicated in the loss of sensitivity of tumorigenic breast cell lines to TGF-beta-mediated growth inhibition. A panel of human breast cell lines, including the immortalized MCF-10F and tumorigenic MCF-7, ZR75-1, BT474, T47-D, MDA-MB231, BT20, and SKBR-3 cell lines, was characterized for responsiveness to TGF-beta-induced G1 growth arrest. Only the nontumorigenic MCF-10F and the tumorigenic MDA-MB231 cell lines demonstrated a significant inhibitory response to TGF-beta1 and a significant binding of 125I-labeled TGF-beta ligand. While expression of TbetaR-I mRNA was similar across the panel of cell lines, TbetaR-II mRNA expression was decreased significantly in all seven tumorigenic cell lines in comparison with the nontumorigenic MCF- 10F cell line. When total cellular protein was fractionated by centrifugation, TbetaR-I protein was observed in both the cytosolic and membrane fractions at similar levels in all cell lines; however, TbetaR-II protein was present in the cytosolic fraction in all cell lines, but was observed in the membrane fraction of only the TGF-beta-responsive MCF-10F and MDA-MB231 cells. Thus, lack of membrane-bound TbetaR-II protein appears to be an important determinant of resistance to TGF-beta-mediated growth inhibition in this group of breast cell lines.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Blotting, Western , Cell Division/drug effects , DNA Mutational Analysis , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , G1 Phase/drug effects , Humans , Mutation/genetics , Protein Serine-Threonine Kinases , Protein Subunits , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
5.
Ann N Y Acad Sci ; 948: 51-66, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11795395

ABSTRACT

Approximately 60% of breast cancer patients have hormone-dependent breast cancer containing estrogen receptors and requiring estrogen for tumor growth. The extent of estrogen biosynthesis and metabolism in the breast cancer tissue microenvironment influences breast-tumor development and growth, and endogenous and exogenous agents may alter the levels of hormonally active estrogens and their metabolites. Isoflavonoid phytoestrogens such as genistein exhibit numerous biochemical activities; however, their effects on estrogen biosynthesis and metabolism in breast cancer cells have not been fully examined. MCF-7 cells (hormone-dependent) and MBA-MB-231 cells (hormone-independent) were treated with genistein (100 nM) for five days and then incubated with radiolabeled estradiol (100 nM, 2.5 microCi) for 0 to 48 h. Media were extracted with ethyl acetate, and the organic residues analyzed by reverse-phase HPLC with a radioactivity flow detector. The major metabolite formed in all cases is estrone, although differences were observed between the cell lines and the various drug treatments. The formation of estrone in untreated MCF-7 cells (approximately 9.3% of radioactivity at 24 h) is relatively limited, in contrast to untreated MDA-MB-231 cells (approximately 32.0% of radioactivity at 24 h). Treatment of MCF-7 cells with 100 nM genistein increased the conversion of estradiol to estrone up to 19.5% in 24 h. The effect of genistein on estrone formation in MDA-MB-231 cells resulted in 37.7% of the radioactivity being estrone. Thus, genistein treatment of breast cancer cells resulted in increased 17-betahydroxysteroid dehydrogenase activity and elevated formation of estrone. Increased levels of oxidative 17-betahydroxysteroid dehydrogenase activity (Type II) were confirmed by Western blots. Therefore, exposure of breast cancer cells to genistein results in elevated conversion of estradiol to estrogenically weaker or inactive metabolites. The regulation of breast-tissue aromatase by exogenous agents such as drugs and environmental agents is being investigated. The benzopyranone-ring system is a molecular scaffold of considerable interest, and this scaffold is found in flavonoid natural products that have weak aromatase inhibitory activity. Medicinal chemistry efforts focus on diversifying the benzopyranone scaffold and utilizing combinatorial chemistry approaches to construct small benzopyranone libraries as potential aro- matase inhibitors. Several compounds in the initial libraries have demonstrated moderate aromatase inhibitory activity in screening assays.


Subject(s)
Aromatase/biosynthesis , Estrogens, Non-Steroidal/pharmacology , Estrogens/biosynthesis , Isoflavones , Antineoplastic Agents, Phytogenic/pharmacology , Aromatase/metabolism , Benzopyrans/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Cell Survival/drug effects , Combinatorial Chemistry Techniques , Estradiol/metabolism , Estrogens/metabolism , Estrogens, Non-Steroidal/chemistry , Female , Humans , Indicators and Reagents , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/prevention & control , Peptide Library , Phytoestrogens , Plant Preparations , Tumor Cells, Cultured
6.
J Steroid Biochem Mol Biol ; 79(1-5): 75-84, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11850210

ABSTRACT

Aromatase (estrogen synthase) is the cytochrome P450 enzyme complex that converts C19 androgens to C18 estrogens. Aromatase activity has been demonstrated in breast tissue in vitro, and expression of aromatase is highest in or near breast tumor sites. Thus, local regulation of aromatase by both endogenous factors as well as exogenous medicinal agents will influence the levels of estrogen available for breast cancer growth. The prostaglandin E2 (PGE2) increases intracellular cAMP levels and stimulates estrogen biosynthesis, and our recent studies have shown a strong linear association between CYP19 expression and the sum of COX-1 and COX-2 expression in breast cancer specimens. PGE2 can bind to four receptor subtypes, EP1-EP4, which are coupled to different intracellular signaling pathways. In primary human breast stromal cell cultures, aromatase activity was significantly induced by PGE2, dexamethasone, and agonists for the EP1 and EP2 receptor subtypes. An EP1 antagonist, SC-19220, inhibited the induction of enzyme activity by PGE2 or 17-phenyltrinor-PGE2, an EP1 agonist. Sulprostone, an EP3 agonist, did not alter aromatase activity levels. Investigations are also underway on the regulation of aromatase by exogenous medicinal agents. Selective steroidal and nonsteroidal agents are effective in inhibiting breast tissue aromatase. The benzopyranone ring system is a molecular scaffold of considerable interest, and this scaffold is found in certain flavonoid natural products that have weak aromatase inhibitory activity. Our novel synthetic route for benzopyranones utilizes readily available salicylic acids and terminal alkynes as starting materials. The synthesis of flavones with diversity on the benzopyranone moiety and at the C-2 position occurs with good to excellent yields using these reaction conditions, resulting in an initial benzopyranone library of thirty compounds exhibiting enhanced and differential aromatase inhibition. Current medicinal chemistry efforts focus on diversifying the benzopyranone scaffold and utilizing combinatorial chemistry approaches to construct small benzopyranone libraries as potential aromatase inhibitors.


Subject(s)
Aromatase Inhibitors , Aromatase/metabolism , Breast/enzymology , Benzopyrans/chemical synthesis , Benzopyrans/chemistry , Benzopyrans/pharmacology , Breast/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Cells, Cultured , Combinatorial Chemistry Techniques , Dinoprostone/pharmacology , Drug Design , Enzyme Induction/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Humans , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/metabolism , Stromal Cells/drug effects , Stromal Cells/enzymology , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Anticancer Res ; 20(1A): 11-9, 2000.
Article in English | MEDLINE | ID: mdl-10769629

ABSTRACT

BACKGROUND: Protein tyrosine phosphatase gamma (PTP gamma) has been implicated as a potential tumor suppressor gene in kidney and lung adenocarcinomas. We have previously shown that PTP gamma mRNA expression levels are lower in DES-induced kidney tumors than in normal kidneys of Syrian hamsters. The goals of the present study were to determine if PTP gamma mRNA is present in both normal and cancerous human breast cells, and to investigate the estrogenic regulation of PTP gamma mRNA expression in these cell types. METHODS: Primary cultured human breast cells derived from surgical specimens of mammoplasty and breast cancer patients, as well as human breast cancer cell lines were used for the study. RT-PCR and RNase protection assay was utilized to detect and quantify levels of PTP gamma mRNA among the cell types used and between control and 17 beta-estradiol (E2)-treated cells. Transient transfection of human estrogen receptor (ER) into MDA-MB-231 human breast cancer cells was performed to establish the role of ER in the regulation of PTP gamma mRNA expression. RESULTS: The results show that PTP gamma mRNA is expressed in primary cultured human breast cells isolated from mammoplasty and breast cancer patients, as well as in human cancer cell lines, and that E2 significantly inhibits PTP gamma expression in ER-positive human breast cancer cells via an ER-mediated mechanism. We show that PTP gamma mRNA levels are lower in human breast cancer cells than in normal human breast cells. Furthermore, we report that PTP gamma mRNA expression is inhibited by E2 in a dose-dependent manner in primary cultured breast cells. After treatment with 20 nM E2 for 24 hours, PTP gamma mRNA was significantly suppressed in primary cultured cancerous and non-cancerous cells from breast cancer patients, as well as in the ER-positive MCF-7 cell line by 50%, 85%, and 66%, respectively. In contrast, the PTP gamma mRNA expression levels did not change in similarly treated ER-negative MDA-MB-231 cells. Sensitivity to E2-induced suppression could be restored (94% inhibition) by transfecting MDA-MB-231 cells with an ER expression plasmid. CONCLUSIONS: Our results are the first to suggest that PTP gamma is a potential estrogen-regulated tumor suppressor gene in human breast cancer which may play an important role in neoplastic processes of human breast epithelium.


Subject(s)
Breast Neoplasms/enzymology , Breast/enzymology , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cells, Cultured , Enzyme Induction , Epithelial Cells/enzymology , Estrogens , Female , Humans , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Nerve Tissue Proteins/genetics , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Receptors, Estrogen/drug effects , Receptors, Estrogen/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Cancer Lett ; 140(1-2): 27-35, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10403538

ABSTRACT

Aromatase, the enzyme system catalyzing estrogen biosynthesis, is found in stromal tissue in the breast. The increased expression of the aromatase CYP19 gene in breast cancer tissues was recently associated with a promoter region regulated through cAMP-mediated pathways. PGE2, derived from cyclooxygenase, increases intracellular cAMP levels and stimulates estrogen biosynthesis. This association suggest that local production of PGE2 via cyclooxgenase isozymes may influence estrogen biosynthesis. The present study represents the first to examine the levels of mRNA expression of CYP19, COX-1, and COX-2 genes in human breast cancer specimens and normal breast tissue samples using semi-quantitative RT-PCR methods. Positive correlations were observed between CYP19 and COX-2 and the greater extent of breast cancer cellularity. Linear regression analysis using a bivariate model shows a strong linear association between CYP19 expression and the sum of COX-1 and COX-2 expression. This significant relationship between the aromatase and cyclooxygenase enzyme systems suggests that autocrine and paracrine mechanisms may be involved in hormone-dependent breast cancer development via growth stimulation from local estrogen biosynthesis.


Subject(s)
Aromatase/biosynthesis , Breast Neoplasms/metabolism , Gene Expression , Prostaglandin-Endoperoxide Synthases/biosynthesis , Breast Neoplasms/pathology , Cell Count , Cyclooxygenase 1 , Cyclooxygenase 2 , Female , Humans , Inflammation/pathology , Isoenzymes/biosynthesis , Linear Models , Membrane Proteins , Reverse Transcriptase Polymerase Chain Reaction
10.
Chem Res Toxicol ; 12(3): 270-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10077490

ABSTRACT

The growth-promoting effects of estrogens in hormone-dependent tumor tissues involve receptor-mediated pathways that are well-recognized; however, the role of estrogens in tumor initiation remains controversial. Estrogen metabolites, primarily the catechol estrogens (CE's), have been implicated in tumor initiation via a redox cycling mechanism. We have developed metabolically stable CE analogues for the study of receptor versus redox cycling effects on DNA damage. Comparisons between hydroxy estradiols (HE2's), methoxy estradiols (ME2's), and hydroxymethyl estradiols (HME2) in potentiometric and DNA damaging studies were made. DNA damage was assessed in calf thymus DNA using 8-oxo-2'-deoxyguanosine (8-oxo-dG) as a genotoxic marker for oxidative stress. Increases in the number of 8-oxo-dG/10(5) dG were significant for each 2-HE2 and 4-HE2. Cu(II)SO4, a transition metal known to catalyze the redox cycling of o-quinones, substantially increased the amount of DNA damage caused by both CE's. However, DNA damage was only observed at concentrations of 10 microM or higher, much greater than what is found under physiologic conditions. Furthermore, the presence of endogenous antioxidants such as glutathione, SOD, and catalase drastically reduced the amount of DNA damage induced by high concentrations of 2-HE2. There was no DNA damage observed for the non-redox cycling HME2's, making these compounds useful probes in the study of receptor-mediated carcinogenesis. Thus, both 2-HE2 and 4-HE2 are capable of producing oxidative DNA damage at micromolar concentrations in vitro. However, since the amount of CE's has not been shown to surpass nanomolar levels in vivo, it is unlikely that free radical production via redox cycling of CE's is a causative factor in human tumorigenesis.


Subject(s)
Carcinogens/toxicity , DNA Damage , Estrogens, Catechol/toxicity , 8-Hydroxy-2'-Deoxyguanosine , Animals , Biomarkers, Tumor , Buffers , Cattle , Chromatography, High Pressure Liquid , Copper/chemistry , DNA/chemistry , Deoxyguanosine/analogs & derivatives , Humans , Lactoperoxidase/chemistry , Oxidation-Reduction , Potentiometry , Reactive Oxygen Species , Spectrophotometry, Ultraviolet
11.
J Steroid Biochem Mol Biol ; 70(4-6): 249-56, 1999.
Article in English | MEDLINE | ID: mdl-10622415

ABSTRACT

Local estradiol production within breast tissue is maintained by the aromatase cytochrome P450arom complex, which has been localized primarily to the stromal component of tumors but also has been detected in the breast epithelial cells. Paracrine interactions between stromal and epithelial components of the breast are critical to the sustained growth and progression of breast tumors. Maintenance of the differentiated state, including hormone and growth factor responsiveness, requires extracellular matrix proteins as substrata for cells. This research has focused on developing a cell culture system that more closely mimics in vivo interactions in order to dissect actual paracrine signaling between these two cell types. Human fibroblasts were isolated from breast tissue and were maintained in a cell culture system grown on plastic support or on a collagen I support matrix. The collagen I matrix model supports cell maintenance and subsequent differentiation on collagen rather than maximal proliferation, therefore allowing for a more accurate environment for the study of hormonal control and cellular communication. Initial experiments compared aromatase activity of patient fibroblasts grown on plastic versus collagen I using the tritiated water release method. Constitutive aromatase activity was found to be lower when cells were grown on a collagen gel for 4-7 days (7.7 fold lower) using DMEM/F12 containing 10% dextran coated charcoal stripped serum. However, fibroblasts grown on collagen I appeared to be significantly more responsive to stimulation by 100 nM dexamethasone (plastic: 6.0 fold induction, collagen: 33.2 fold induction) when pretreated for 12 h prior to measurement of aromatase activity. In an effort to examine paracrine interactions between the stromal and epithelial cells in breast tissue, experiments using conditioned media from fibroblast cultures were performed. Testosterone administration to fibroblasts results in the production of estradiol into the media in sufficient concentrations to elicit an increase in pS2 expression when the conditioned media is administered to MCF-7 cells. The addition of a potent aromatase inhibitor resulted in a complete suppression of fibroblast-derived estrogens and showed only a modest increase in pS2 expression. Culturing breast fibroblasts and epithelial cells on extracellular matrix allows for a more meaningful examination of the paracrine interactions between these cell types within the context of an appropriate extracellular environment. This study highlights the need for evaluation of gene expression in cell culture systems that accurately reflect the tissue microenvironment.


Subject(s)
Aromatase/metabolism , Breast Neoplasms/enzymology , Breast/cytology , Breast/enzymology , Collagen/pharmacology , Adenocarcinoma/enzymology , Breast/drug effects , Cell Division , Cells, Cultured , Dexamethasone/pharmacology , Epithelial Cells/physiology , Extracellular Matrix , Female , Gene Expression Regulation/drug effects , Humans , RNA, Messenger/genetics , Stromal Cells/drug effects , Stromal Cells/enzymology , Testosterone/pharmacology , Thymidine/metabolism , Tumor Cells, Cultured , Vimentin/genetics
12.
Int J Oncol ; 13(6): 1291-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9824647

ABSTRACT

We report here that (-)-gossypol significantly inhibits the proliferation of stromal cells derived from human breast adipose tissues (human breast adipose stromal cells) in a dose-dependent manner. The mechanisms involved in the anti-proliferative action of (-)-gossypol on adipose stromal cells were also investigated. (-)-Gossypol stimulated transforming growth factor (TGFbeta1) secretion after 24 h, and RNase protection assay showed that TGFbeta1 mRNA levels were increased as well. We also observed that TGFbeta1 significantly inhibited the growth of human breast adipose stromal cells in a dose-dependent manner. When human breast adipose stromal cells were co-incubated with 5 microM (-)-gossypol and 50 microgram/ml of anti-TGF-beta1,-beta2,-beta3 antibody, growth inhibition caused by (-)-gossypol was completely abrogated. This study indicates that the anti-proliferative activity of (-)-gossypol on human breast adipose stromal cells may be mediated by changes in TGF 1 production.


Subject(s)
Adipose Tissue/drug effects , Breast/drug effects , Gossypol/pharmacology , Transforming Growth Factor beta/biosynthesis , Adipose Tissue/cytology , Adult , Antibodies/immunology , Breast/cytology , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , DNA/drug effects , Female , Humans , Middle Aged , RNA, Messenger/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/pharmacology
13.
Anticancer Res ; 18(4A): 2541-6, 1998.
Article in English | MEDLINE | ID: mdl-9703907

ABSTRACT

Estrogen is the major hormonal stimulus for growth of the hormonal-dependent type of breast cancer. The rate-limiting step in the conversion of androgens to estrogens in breast tumors is catalyzed by aromatase, one of a series of related P-450 enzymes involved in the production of steroid hormones. An interesting correlation has been found between KGF mRNA and aromatase mRNA expression in human breast tumors. Tumors that express aromatase mRNA exhibit strong KGF expression, while tumors that do not express aromatase are weak or negative for KGF expression. Thus, it is reasonable to theorize that a possible association between KGF and aromatase in controlling human breast tumor growth exists. The purpose of the current study was to establish whether there is any interaction between KGF, which is known to have epithelial-specific mitogenic activity on breast cancer cells in vitro, and the synthesis of estradiol within the hormone-dependent breast cancer epithelial cells. In the present study, we have demonstrated that KGF stimulates aromatase activity in human breast cancer cells (MCF-7) in a dose-dependent manner. Our data shows that recombinant human KGF, at a dose as low as 10 ng/ml, can significantly increase aromatase activity 2-fold over controls. In agreement with this observation, we also found that aromatase mRNA levels were increased after 10 ng/ml KGF treatment in MCF-7 cells. These results indicate that the stimulatory effect of KGF on aromatase activity may be mediated by alterations in aromatase mRNA levels or in the efficiency of the translation of the message in MCF-7 cells. In addition, our results have demonstrated that modulation of aromatase activity appears to correlate with the stimulation of proliferative activity by KGF in MCF-7 cells. These results are consistent with our previous observations that estradiol-17 beta stimulates KGF expression in human breast cancer stromal cells, leading to the speculation that breast malignant transformation is associated with a positive feedback stimulation, whereby estradiol-17 beta stimulates breast cancer stromal cell production of KGF, and KGF subsequently stimulates aromatase activity in breast cancer cells, consequently raising levels of estradiol-17 beta, in turn acts on breast stromal cells to yield more KGF. Such a positive feedback loop could play an important role in the loss of growth control in human breast cancer cells.


Subject(s)
Aromatase/biosynthesis , Breast Neoplasms/enzymology , Fibroblast Growth Factors , Growth Substances/pharmacology , Aromatase/metabolism , Cell Division , Enzyme Induction , Estradiol/metabolism , Female , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kinetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Recombinant Proteins/pharmacology , Testosterone/metabolism , Transcription, Genetic/drug effects , Tumor Cells, Cultured
14.
Oncol Rep ; 5(3): 577-83, 1998.
Article in English | MEDLINE | ID: mdl-9538155

ABSTRACT

The local recurrence rate of breast cancer has been reported to be unusually high at the surgical scar. Such breast cancer recurrence is believed to be triggered by the release of growth factors into the healing wound. Observations from an animal model have also demonstrated that KGF expression is dramatically induced by creation of full thickness wounds in mouse skin. Since KGF is an epithelial cell-specific mitogen in rat mammary epithelium, it is reasonable to speculate that KGF may be also involved in regulating human breast cancer cell growth. The purpose of the present study was to determine the effect of estradiol-17 on KGF gene expression in normal human breast stromal cells, as well as in human breast cancer stromal cells, and the mechanisms by which estradiol-17 regulates breast epithelial proliferation. Our results show that KGF expression was not effected by estradiol-17 treatment in normal human breast stromal cells. In contrast, KGF expression was stimulated by estradiol-17 in human breast cancer stromal cells. KGF mRNA levels have also been examined in normal human breast stromal cells and human breast cancer stromal cells. An interesting correlation was found between KGF expression and estradiol-17 regulation in these cell types. Normal human breast stromal cells which do not response to estradiol-17 have lower KGF mRNA level than the cancer cells which KGF expression is stimulated by estradiol-17. Our data also demonstrate that recombinant human KGF significantly stimulate normal human breast and human breast cancer epithelial cell proliferation in a dose-dependent manner. Since we have shown that estradiol-17 induces KGF mRNA expression in human breast cancer stromal cells, KGF may be involved at least in part in the stimulatory pathway that is initiated by estradiol-17 in human breast cancer epithelial cells.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Carcinoma, Ductal, Breast/metabolism , Estradiol/pharmacology , Fibroblast Growth Factors , Growth Substances/metabolism , RNA, Messenger/metabolism , Breast/cytology , Breast/drug effects , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , DNA Primers/chemistry , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estradiol/administration & dosage , Female , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Growth Substances/genetics , Humans , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Cells, Cultured/drug effects
15.
J Med Chem ; 40(23): 3756-64, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9371241

ABSTRACT

A series of synthetic estrogens containing hydroxyalkyl side chains at the C-4 position of the A ring were designed as metabolically stable analogs of 4-hydroxyestradiol, a catechol estrogen. These synthetic steroids would facilitate investigations on the potential biological role of catechol estrogens and also enable further examination of the structural and electronic constraints on the A ring in the interaction of estrogens with the estrogen receptor. Catechol estrogens are implicated as possible causative agents in estrogen-induced tumorigenesis. 4-Hydroxyestradiol has weaker affinity for the estrogen receptor and exhibits lower estrogenic activity in vivo; on the other hand, the catechol estrogens are prone to further oxidative metabolism and can form reactive intermediates. This report describes the synthesis and initial biochemical evaluation of 4-(hydroxyalkyl)estrogens and 4-(aminoalkyl)estradiols. The 4-(hydroxyalkyl)estrogens were prepared by oxidative hydroboration of 4-alkenylestradiols. The alkenylestradiols were obtained via a Stille cross-coupling between a MOM-protected 4-bromoestradiol and an alkenylstannane. The (4-aminoalkyl)estrogens were prepared from the hydroxyalkyl derivatives with phthalimide under Mitsunobu conditions. The substituted estradiols were evaluated for estrogen receptor binding activity in MCF-7 human mammary carcinoma cells, and 4-(hydroxymethyl)estradiol 1 exhibited the highest affinity with an apparent EC50 value of 364 nM. The relative activities for mRNA induction of the pS2 gene in MCF-7 cell cultures by the 4-(hydroxyalkyl)estrogens closely parallel the relative binding affinities. 4-(Hydroxymethyl)estradiol 1 did not stimulate the growth of MCF-7 cells at concentrations up to 1 microM. Thus, 4-(hydroxymethyl)estradiol 1 exhibited similar estrogen receptor affinity as the catechol estrogen, 4-hydroxyestradiol, and may prove useful in the examination of the biological effects of 4-hydroxyestrogens.


Subject(s)
Estradiol/analogs & derivatives , Estrogens, Catechol/chemical synthesis , Estrogens, Catechol/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/ultrastructure , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/ultrastructure , Cell Division/drug effects , Estradiol/chemical synthesis , Estradiol/pharmacokinetics , Estradiol/pharmacology , Humans , Kinetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/ultrastructure , Receptors, Estrogen/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured/drug effects
16.
Breast Cancer Res Treat ; 44(1): 57-64, 1997 May.
Article in English | MEDLINE | ID: mdl-9164678

ABSTRACT

The aromatase cytochrome P450 complex is responsible for the in vivo conversion of androgens to estrogens. Although breast cancer epithelial cells have been reported to have appreciable aromatase activity, its biologic significance remains uncertain. To address this, the effect of androgens on the expression of the estrogen-regulated gene pS2 in hormone-dependent human breast carcinoma cells in vitro was examined. Steroid-deprived MCF-7 cells were exposed to varying concentrations (1 nM, 10 nM, and 100 nM of androstenedione or testosterone for 2,4, and 6 days. Baseline aromatase activity was 4.9 (+/-3.1) fmol 3H2O/hour/microgram DNA [34.3 (+/-21.3) fmol/hr/10(6) cells] and was not influenced by the androgens. As an indication of estrogen biosynthesis, northern analysis was performed to quantitate pS2 mRNA expression. Although no significant pS2 induction was observed at 2 days, both 4 and 6 day exposure to 100 nM testosterone resulted in a 3-fold increase in pS2 mRNA expression. 5 alpha-dihydrotestosterone (5 alpha-DHT) failed to elicit a similar pS2 response. This testosterone-induced response was inhibited with the aromatase inhibitor 7 alpha (4'-amino) phenylthio-1,4-androstadiene-3,17-dione (7 alpha-APTADD) and with 10 microM tamoxifen. MCF-7 breast cancer cells possess endogenous aromatase activity at high enough levels to convert androgens to estrogens and elicit an estrogen-induced response. The expression of aromatase may offer a potential advantage to hormone-responsive cells, providing an additional autocrine growth pathway which may be exploited.


Subject(s)
Androgens/pharmacology , Aromatase/pharmacology , Breast Neoplasms/pathology , Estrogens/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Dose-Response Relationship, Drug , Estrogens/pharmacology , Female , Humans , Protein Biosynthesis , Proteins/drug effects , Trefoil Factor-1 , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Suppressor Proteins
17.
J Steroid Biochem Mol Biol ; 61(1-2): 73-7, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9328212

ABSTRACT

The JAr choriocarcinoma cell cultures have demonstrated high levels of aromatase activity and have been useful for assaying a wide variety of aromatase inhibitors for aromatase inhibition in intact cells. Recently, several 7alpha-arylaliphatic androgens have shown effective inhibition of human placental microsomal aromatase in vitro, with apparent Ki values ranging from 10 to 20 nM. A series of 7alpha-arylaliphatic androst-4-ene-3,17-dione compounds demonstrated potent competitive inhibition, and 7alpha-arylaliphatic androsta-1,4-diene-3,17-diones were enzyme-activated irreversible inhibitors. Both series of these potent inhibitors were investigated for the ability to inhibit aromatase activity in JAr cells by measuring the conversion of [1beta-3H]-androstenedione to 3H2O and unlabelled estrone. JAr cell cultures were incubated for 2 h at 37 degrees C with the aromatase inhibitors at concentrations of 10 pM to 10 microM, the percentage of enzyme inhibition was determined, and IC50 values for inhibitors were calculated. Both series of synthetic compounds demonstrated good to excellent aromatase inhibition, and the most effective inhibitors in both series were those compounds with a phenylpropyl substituent at the 7alpha-position of the steroid nucleus. The 7alpha-arylaliphatic androst-4-ene-3,17-diones exhibited inhibition of JAr aromatase activity with IC50 values from 300 to 434 nM. More potent aromatase inhibition was observed with the 7alpha-arylaliphatic androsta-1,4-diene-3,17-diones, which exhibited IC50 values from 64 to 232 nM. Enhanced efficacy of steroidal enzyme-activated irreversible inhibitors compared to competitive inhibitors was observed in these studies and is consistent with previous reports. These results suggest that JAr choriocarcinoma cells with high levels of aromatase activity may be useful in differentiating steroidal aromatase inhibitors exhibiting different mechanisms of enzyme inhibition. In summary, the 7alpha-phenylpropyl androsta-1,4-diene-3,17-dione analogs, which are enzyme-activated irreversible inhibitors, demonstrated the most effective inhibition of aromatase activity present in the JAr cell cultures among the various 7alpha-arylaliphatic androgens.


Subject(s)
Androstenedione/analogs & derivatives , Androstenedione/pharmacology , Aromatase Inhibitors , Choriocarcinoma/enzymology , Testosterone Congeners/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Tumor Cells, Cultured
18.
J Steroid Biochem Mol Biol ; 61(3-6): 247-54, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9365197

ABSTRACT

The inhibition of aromatase, the enzyme responsible for converting androgens to estrogens, is therapeutically useful for the endocrine treatment of hormone-dependent breast cancer. Research by our laboratory has focused on developing competitive and irreversible steroidal aromatase inhibitors, with an emphasis on synthesis and biochemistry of 7alpha-substituted androstenediones. Numerous 7alpha-thiosubstituted androst-4-ene-3,17-diones are potent competitive inhibitors, and several 1,4-diene analogs, such as 7alpha-(4'-aminophenylthio)-androsta-1,4-diene-3,17-di one (7alpha-APTADD), have demonstrated effective enzyme-activated irreversible inhibition of aromatase in microsomal enzyme assays. One focus of current research is to examine the effectiveness and biochemical pharmacology of 7alpha-APTADD in vivo. In the hormone-dependent 7,12-dimethylbenz(a)anthracene (DMBA)-induced rat mammary carcinoma model system, 7alpha-APTADD at a 50 mg/kg/day dose caused an initial decrease in mean tumor volume during the first week, and tumor volume remained unchanged throughout the remaining 5-week treatment period. This agent lowers serum estradiol levels and inhibits ovarian aromatase activity. A second research area has focused on the synthesis of more metabolically stable inhibitors by replacing the thioether linkage at the 7alpha position with a carbon-carbon linkage. Several 7alpha-arylaliphatic androst-4-ene-3,17-diones were synthesized by 1,6-conjugate additions of appropriate organocuprates to a protected androst-4,6-diene or by 1,4-conjugate additions to a seco-A-ring steroid intermediate. These compounds were all potent inhibitors of aromatase with apparent Kis ranging between 13 and 19 nM. Extension of the research on these 7alpha-arylaliphatic androgens includes the introduction of a C1-C2 double bond in the A-ring to provide enzyme-activated irreversible inhibitors. The desired 7alpha-arylaliphatic androsta-1,4-diene-3,17-diones were obtained from their corresponding 7alpha-arylaliphatic androst-4-ene-3,17-diones by oxidation with 2,3-dichloro-5,6-dicyano-1,4-benzoquinone (DDQ). These inhibitors demonstrated enzyme-mediated inactivation of aromatase with apparent k(inact)s ranging from 4.4 x 10(-4) to 1.90 x 10(-3) s(-1). The best inactivator of the series was 7alpha-phenpropylandrosta-1,4-diene-3,17-dione, which exhibited a T(1/2) of 6.08 min. Aromatase inhibition was also observed in MCF-7 human mammary carcinoma cell cultures and in JAr human choriocarcinoma cell cultures, exhibiting IC50 values of 64-328 nM. The 7alpha-arylaliphatic androgens thus demonstrate potent inhibition of aromatase in both microsomal incubations and in choriocarcinoma cell lines expressing aromatase enzymatic activity. Additionally, the results from these studies provide further evidence for the presence of a hydrophobic binding pocket existing near the 7alpha-position of the steroid in the active site of aromatase. The size of the 7alpha-substituent influences optimal binding of steroidal inhibitors to the active site and affects the extent of enzyme-mediated inactivation observed with androsta-1,4-diene-3,17-dione analogs.


Subject(s)
Androstenedione/chemistry , Androstenedione/pharmacology , Aromatase Inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Androstenedione/analogs & derivatives , Animals , Breast Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Female , Humans , Rats
19.
Anticancer Res ; 17(2A): 1003-9, 1997.
Article in English | MEDLINE | ID: mdl-9137441

ABSTRACT

Recently we demonstrated that gossypol (GP), a male antifertility agent, is a potent inhibitor of malignant human prostate cancer cell growth that acts by arresting cells in G0/G1 phase and that this inhibitory effect may be mediated by transforming growth factor-beta 1 (TGF-beta 1). In this study we examined the effect of GP on the growth of prostatic cells from human benign prostatic hyperplasia (BPH) patients in vitro. Consistent with its inhibitory effect on the growth of malignant human prostate cancer cells, GP also acts as a potent inhibitor of cultured human BPH cell growth as assessed by thymidine incorporation assay. These results were confirmed by flow cytometric analysis which revealed that treatment of human BPH cells with increasing concentrations of GP resulted in a dose-dependent accumulation of cells in the G0/G1 phase with a concomitant decrease in cells progressing to the S and G2/M phases. Since inhibition of prostate cancer cells by GP appears to be mediated by TGF-beta 1, we also investigated the effect of GP on TGF-beta 1 gene expression in BPH cells. The results show that GP treatment resulted in a marked elevation of TGF-beta 1 gene expression indicating that TGF-beta 1 might be involved at least in part in the inhibitory pathway that is initiated by GP.


Subject(s)
Gossypol/therapeutic use , Prostatic Hyperplasia/drug therapy , Cell Division/drug effects , G1 Phase/drug effects , Humans , Male , Prostatic Hyperplasia/pathology , RNA, Messenger/analysis , Resting Phase, Cell Cycle/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
20.
Cancer Lett ; 107(1): 37-44, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8913264

ABSTRACT

Gossypol (GP), an antifertility agent in males, is also capable of inhibiting the proliferation of a wide range of cancer cells in vivo and in vitro. Thus, in this study we investigated the effect of GP on the growth of human androgen-independent prostate cancer cell line (PC3). The results showed that GP acts as a potent inhibitor of PC3 cells as determined by thymidine incorporation assay and flow cytometric analysis. Flow cytometry revealed that treatment of PC3 cells with GP resulted in a dose- and time-dependent accumulation of cells in the GO/GI phase with a concomitant decrease in cells progressing to the S and G2/M phase. These data support our thymidine incorporation results which indicated that GP is a potent inhibitor of PC3 cells. By ribonuclease protection assay, we also investigated the effect of GP on transforming growth factor-beta 1 (TGF-beta 1) gene expression in PC3 cells. Interestingly, the stimulatory effect of GP on TGF-beta 1 gene expression correlates well with its inhibitory effect on PC3 cell DNA synthesis and its ability to arrest cells in GO/G1 phase. Based on these data, it can be concluded that GP is a potent inhibitor of prostate cancer cell growth that acts by arresting cells in GO/G1 phase and that this inhibitory effect may be mediated by TGF-beta 1.


Subject(s)
Cell Division/drug effects , Gossypol/pharmacology , Prostatic Neoplasms/pathology , Transforming Growth Factor beta/biosynthesis , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Humans , Male , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...