Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Cell Oncol (Dordr) ; 44(1): 205-218, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33119860

ABSTRACT

PURPOSE: Fibroblast growth factor receptors (FGFR) and pathways are important players in breast cancer (BC) development. They are commonly altered, and BCs exhibiting FGFR gene amplification are currently being studied for drug development. Here, we aimed to compare the effects of three FGFR inhibitors (FGFRis), i.e., non-selective TKI258 and selective BGJ398 and AZD4547, on different BC-derived cell lines (BCCs) and primary tissues. METHODS: The human BCCs MCF-7 and MDA-MB-231(SA) (wild-type FGFR) and MFM223 (amplified FGFR1 and FGFR2) were analyzed for FGFR expression using qRT-PCR, and the effects of FGFRis on FGFR signaling by Western blotting. The effects of FGFRis on proliferation, viability, migration and invasion of BCCs were assessed in 2D cultures using live-cell imaging, and in 3D cultures using phenotypic analysis of organoids. To study radio-sensitization, FGFRi treatment was combined with irradiation. Patient-derived BC samples were treated with FGFRis in explant cultures and immunostained for Ki67 and cleaved caspase 3. RESULTS: We found that all FGFRis tested decreased the growth and viability of BC cells in 2D and 3D cultures. BGJ398 and AZD4547 were found to be potent at low concentrations in FGFR-amplified MFM233 cells, whereas higher concentrations were required in non-amplified MCF7 and MDA-MB-231(SA) cells. TKI258 inhibited the migration and invasion, whereas BGJ398 and AZD4547 only inhibited the invasion of MDA-MB-231(SA) cells. FGFRi treatment of MCF7 and MFM223 cells enhanced the inhibitory effect of radiotherapy, but this effect was not observed in MDA-MB-231(SA) cells. FGFRi-treated primary BC explants with moderate FGFR levels showed a tendency towards decreased proliferation and increased apoptosis. CONCLUSIONS: Our results indicate that, besides targeting FGFR-amplified BCs with selective FGFRis, also BCs without FGFR amplification/activation may benefit from FGFRi-treatment. Combination with other treatment modalities, such as radiotherapy, may allow the use of FGFRis at relatively low concentrations and, thereby, contribute to better BC treatment outcomes.


Subject(s)
Benzamides/pharmacology , Benzimidazoles/pharmacology , Breast Neoplasms/pathology , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolones/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Tissue Culture Techniques , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Neoplasm Invasiveness , Organoids/drug effects , Organoids/pathology , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects
2.
Mol Cell Endocrinol ; 461: 194-204, 2018 02 05.
Article in English | MEDLINE | ID: mdl-28923346

ABSTRACT

Fibroblast growth factors (FGF) and their receptors (FGFRs) regulate many developmental processes including differentiation of mesenchymal stromal cells (MSC). We developed two MSC lines capable of differentiating to osteoblasts and adipocytes and studied the role of FGFRs in this process. We identified FGFR2 and fibroblast growth factor receptor like-1 (FGFRL1) as possible actors in MSC differentiation with gene microarray and qRT-PCR. FGFR2 and FGFRL1 mRNA expression strongly increased during MSC differentiation to osteoblasts. FGF2 treatment, resulting in downregulation of FGFR2, or silencing FGFR2 expression with siRNAs inhibited osteoblast differentiation. During adipocyte differentiation expression of FGFR1 and FGFRL1 increased and was down-regulated by FGF2. FGFR1 knockdown inhibited adipocyte differentiation. Silencing FGFR2 and FGFR1 in MSCs was associated with decreased FGFRL1 expression in osteoblasts and adipocytes, respectively. Our results suggest that FGFR1 and FGFR2 regulate FGFRL1 expression. FGFRL1 may mediate or modulate FGFR regulation of MSC differentiation together with FGFR2 in osteoblastic and FGFR1 in adipocytic lineage.


Subject(s)
Adipocytes/cytology , Cell Differentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Down-Regulation/drug effects , Fibroblast Growth Factor 2/pharmacology , Gene Expression Profiling , Gene Silencing , Male , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptors, Fibroblast Growth Factor/genetics
3.
Horm Metab Res ; 46(5): 328-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24526372

ABSTRACT

Ospemifene is a tissue-selective estrogen agonist/antagonist that was recently approved for the treatment of dyspareunia associated with vulvar and vaginal atrophy, which occurs in up to approximately 50% of postmenopausal women. The current analyses were conducted to determine whether ospemifene exhibits estrogenic activity in the mammary glands of ovariectomized rats and to compare potential estrogenic activity with selective estrogen receptor modulators (tamoxifen, raloxifene, and toremifene). Three separate studies with differing durations (6, 9, and 28 days) were conducted using similar procedures in ovariectomized Sprague-Dawley rats. Estradiol treatment and sham-treated ovariectomized rats were used as positive and negative controls, respectively. Cell proliferation was examined using labeled 5-bromo-2-deoxyuridine; cytoplasmic prolactin was characterized with antibody staining. The morphology of the mammary gland was studied by histological staining of sections from the right fourth mammary glands, and the excised gland from the left side was used for counting the lobulus number. Neither ospemifene nor selective estrogen receptor modulators substantially induced 5-bromo-2-deoxyuridine staining, altered the morphology of the mammary glands, or changed prolactin immunostaining in ovariectomized rats compared with the ovariectomized controls. With the exception of toremifene, the selective estrogen receptor modulators did not cause a substantial induction in mammary gland lobuli. Estradiol had effects opposite to those of the selective estrogen receptor modulators in these studies. Ospemifene exhibited no substantial estrogenic activity in the mammary gland of ovariectomized rats. Activity in the mammary gland of ovariectomized rats with ospemifene was comparable to raloxifene and tamoxifen.


Subject(s)
Cell Proliferation/drug effects , Estrogen Antagonists/pharmacology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/physiology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/analogs & derivatives , Animals , Female , Mammary Glands, Animal/anatomy & histology , Ovariectomy , Rats , Rats, Sprague-Dawley , Tamoxifen/pharmacology
4.
Horm Metab Res ; 46(1): 27-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24108389

ABSTRACT

Ospemifene is a non-estrogen agent that exerts tissue-specific estrogen agonistic and weak antagonistic effects (i. e., is a selective estrogen receptor modulator [SERM]). The effects of various once-daily oral doses of ospemifene on bone are examined across 3 studies for 4 or 52 weeks after surgery in the ovariectomized (OVX) rat model of postmenopausal bone loss. Ospemifene treatment reduced the loss of bone mineral content and density observed in untreated OVX rats, significantly increased distal femur bone mineral content at 51 weeks at 25 mg/kg dose compared with untreated OVX rats (p<0.01), and significantly increased trabecular bone mineral density of the distal femur and proximal tibia with 1, 5, or 25 mg/kg doses after 52 weeks. Ospemifene 5 and 25 mg/kg preserved distal femur trabecular structure; trabecular number was significantly increased, whereas trabecular separation and eroded surface values were significantly decreased (all p<0.01). Structural changes associated with ospemifene were accompanied by increased mechanical strength of femurs and 4th lumbar vertebra compared with untreated OVX rats. Ospemifene 10 mg/kg prevented OVX-induced bone loss; trabecular bone volume of distal femurs was increased after 4 weeks. Further, histomorphometric measures revealed decreased bone resorption after 4 weeks of ospemifene treatment, with effects similar to other SERMs (raloxifene and droloxifene). Ospemifene 3 and 10 mg/kg significantly inhibited OVX-induced increases in osteoclast number, and doses ≥0.3 mg/kg dose-dependently reversed the OVX-induced increase in the double-labeled volume:bone volume ratio. These results demonstrate antiresorptive, selective agonist effects of ospemifene on bone that appear similar to raloxifene in this in vivo animal model of estrogen deficiency.


Subject(s)
Bone and Bones/drug effects , Bone and Bones/physiology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/analogs & derivatives , Animals , Biomechanical Phenomena/drug effects , Body Weight/drug effects , Bone Density/drug effects , Bone Remodeling/drug effects , Bone and Bones/anatomy & histology , Cell Count , Compressive Strength/drug effects , Female , Femur/anatomy & histology , Femur/drug effects , Femur/physiology , Lumbar Vertebrae/anatomy & histology , Lumbar Vertebrae/drug effects , Lumbar Vertebrae/physiology , Luteinizing Hormone/blood , Organ Size/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley , Tamoxifen/pharmacology , Tibia/anatomy & histology , Tibia/drug effects , Tibia/physiology , Uterus/drug effects , Uterus/metabolism
5.
Mol Cell Endocrinol ; 358(1): 104-15, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22465097

ABSTRACT

Fibroblast growth factor-8 (FGF-8) is implicated in the development and progression of breast cancer and its levels are frequently elevated in breast tumors. The mechanisms driving FGF-8-mediated tumorigenesis are not well understood. Herein we aimed to identify target genes associated with FGF-8b-mediated breast cancer cell proliferation by carrying out a cDNA microarray analysis of genes expressed in estrogen receptor negative S115 breast cancer cells treated with FGF-8b for various time periods in comparison with those expressed in non-treated cells. Gene and protein expression was validated for selected genes by qPCR and western blotting respectively. Furthermore, using TRANSBIG data, the expression of human orthologs of FGF-8-regulated genes was correlated to the Nottingham prognostic index and estrogen receptor status. The analysis revealed a number of significantly up- and down-regulated genes in response to FGF-8b at all treatment times. The most differentially expressed genes were genes related to cell cycle regulation, mitosis, cancer, and cell death. Several key regulators of early cell cycle progression such as Btg2 and cyclin D1, as well as regulators of mitosis, including cyclin B, Plk1, survivin, and aurora kinase A, were identified as novel targets for FGF-8b, some of which were additionally shown to correlate with prognosis and ER status in human breast cancer. The results suggest that in stimulation of proliferation FGF-8b not only promotes cell cycle progression through the G1 restriction point but also regulates key proteins involved in chromosomal segregation during mitosis and cytokinesis of breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle/genetics , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Mammary Neoplasms, Animal/genetics , Mitosis/genetics , Animals , Apoptosis/genetics , Aurora Kinase A , Aurora Kinases , Breast Neoplasms/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Cyclin B/genetics , Cyclin B/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Fibroblast Growth Factor 8/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mammary Neoplasms, Animal/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering , Receptors, Estrogen/biosynthesis , Signal Transduction , Survivin , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Polo-Like Kinase 1
6.
Acta Neurol Scand ; 122(1): 63-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19951275

ABSTRACT

OBJECTIVE: We examined the prevalence and associated factors of restless legs syndrome (RLS) in a 57-year-old unselected urban population in northern Finland. METHODS: A health survey was conducted in 2002 that targeted persons born in 1945 and residing in the city of Oulu on 31 December, 2001. Their history of RLS, coronary heart disease (CHD), daytime sleepiness, depressive symptoms and snoring was assessed by means of questionnaires. RESULTS: Altogether 995 of 1332 eligible subjects (74%) participated (556 women, 439 men). The overall prevalence of RLS > or = 1 per week was 20% in women and 15% in men. In the fitted multiple logistic regression model, RLS was found to be associated with female gender (OR 1.64, 95% CI 0.98-2.72), CHD (OR 2.92, 95% CI 1.18-7.23), daytime sleepiness (OR 2.12, 95% CI 1.32-3.41), moderately elevated (31-45) or high (46-65) Zung sum scores (OR 1.95, 95% CI 1.09-3.48 and OR 3.67, 95% CI 1.71-7.90, respectively), antidepressant medication (OR 2.10, 95% CI 1.06-4.19) and arthropathy (OR 1.69, 95% CI 1.04-2.72). Insufficient evidence was found of an association between RLS and type 2 diabetes or impaired glucose regulation. CONCLUSIONS: Restless legs syndrome is fairly common in subjects aged 57 years. A particularly strong positive association was observed between RLS and depressive symptoms and CHD.


Subject(s)
Restless Legs Syndrome/epidemiology , Coronary Disease/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Female , Finland/epidemiology , Glucose Tolerance Test , Humans , Logistic Models , Male , Middle Aged , Prevalence , Restless Legs Syndrome/diagnosis
7.
Br J Pharmacol ; 151(3): 384-95, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17420779

ABSTRACT

BACKGROUND AND PURPOSE: Several selective oestrogen receptor modulators (SERMs) with oestrogen agonist effects in bone cells and without increased risk of breast and endometrial cancer have been developed. Here, we have investigated the effects of different types of SERMs on osteoclast differentiation, bone resorption and apoptosis in vitro. EXPERIMENTAL APPROACH: Human peripheral blood-derived CD14+ monocytes were cultured on bovine bone slices in the presence of RANKL, M-CSF, TNF-alpha and dexamethasone for seven days. Also, CD14+ monocytes were co-cultured either with human SaOS-2 or MG-63 osteosarcoma cells, in the presence of parathyroid hormone. Osteoclast cultures were treated with different SERMs. TRACP+ multinucleated cells and C-terminal telopeptide of type I collagen were used as markers for osteoclast formation and bone resorption, respectively. KEY RESULTS: In CD14+ monocyte cultures, tamoxifen directly inhibited human osteoclast formation and bone resorption, while raloxifene and ospemifene had no inhibitory effect. In the co-cultures either with SaOS-2 or MG-63 cells, ospemifene and raloxifene as well as tamoxifen inhibited osteoclast formation in a concentration-dependent manner. The inhibitory effect was associated with an increased production of osteoprotegerin. The anti-oestrogen ICI 182 780 completely reversed the effects of these SERMs. CONCLUSION AND IMPLICATIONS: Tamoxifen had an oestrogen receptor dependent, direct, inhibitory effect on human osteoclast differentiation and bone resorption, whereas ospemifene and raloxifene required osteoblastic cells to achieve a similar inhibition. The effects of ospemifene and raloxifene were mediated by oestrogen receptors by a mechanism involving paracrine induction of osteoprotegerin in cultures with osteoblast derived osteosarcoma cells.


Subject(s)
Osteoclasts/drug effects , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Bone Resorption/prevention & control , Cattle , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Fulvestrant , Humans , Lipopolysaccharide Receptors/analysis , Macrophage Colony-Stimulating Factor/pharmacology , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteoprotegerin/biosynthesis , RANK Ligand/pharmacology , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
9.
Mol Cell Endocrinol ; 248(1-2): 141-8, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16406264

ABSTRACT

Sex steroid hormone signaling regulates the development, growth, and functioning of the breast and the prostate and plays a role in the development and progression of cancer in these organs. The intracellular concentration of active sex steroid hormones in target tissues is regulated by several enzymes, including 17beta-hydroxysteroid dehydrogenases (17HSDs). Changes in the expression patterns of these enzymes may play a pathophysiological role in malignant transformation. We recently analyzed the mRNA expressions of the 17HSD type 1, 2, and 5 enzymes in about 800 breast carcinoma specimens. Both 17HSD type 1 and 2 mRNAs were detected in normal breast tissue from premenopausal women but not in specimens from postmenopausal women. The patients with tumors expressing 17HSD type 1 mRNA or protein had significantly shorter overall and disease-free survival than the other patients. The expression of 17HSD type 5 was significantly higher in breast tumor specimens than in normal tissue. Cox multivariate analyses showed that 17HSD type 1, tumor size, and estrogen receptor alpha (ERalpha) had independent prognostic significance. We developed, using a LNCaP prostate cancer cell line, a model to study the malignant transformation of prostate cancer and showed that androgen-sensitive LNCaP cells are transformed into neuroendocrine-like cells when cultured without androgens and, eventually into highly proliferating androgen-independent cells. We conducted Northern hybridizations and microarrays to analyze the gene expression during these processes. Substantial changes in the expressions of steroid metabolizing enzymes occurred during the transformation process. The variations in steroid-metabolizing enzymes during cancer progression may be crucial in the regulation of the growth and function of organs.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , Breast Neoplasms/enzymology , Cell Proliferation , Gonadal Steroid Hormones/metabolism , Prostatic Neoplasms/enzymology , 17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/genetics , Androgens/metabolism , Breast Neoplasms/pathology , Female , Humans , Male , Prostate/enzymology , Prostate/metabolism
10.
Apoptosis ; 10(6): 1395-410, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16215679

ABSTRACT

Tamoxifen (Tam) is widely used in chemotherapy of estrogen receptor-positive breast cancer. It inhibits proliferation and induces apoptosis of breast cancer cells by estrogen receptor-dependent modulation of gene expression, but recent reports have shown that Tam (especially at pharmacological concentrations) has also rapid nongenomic effects. Here we studied the mechanisms by which Tam exerts rapid effects on breast cancer cell viability. In serum-free medium 5-7 microM Tam induced death of MCF-7 and MDA-MB-231 cells in a time-dependent manner in less than 60 min. This was associated with release of mitochondrial cytochrome c, a decrease of mitochondrial membrane potential and an increase in production of reactive oxygen species (ROS). This suggests that disruption of mitochondrial function has a primary role in the acute death response of the cells. Accordingly, bongkrekic acid, an inhibitor of mitochondrial permeability transition, was able to protect MCF-7 cells against Tam. Rapid cell death induction by Tam was not associated with immediate activation of caspase-9 or cleavage of poly (ADP-ribose) polymerase. It was not blocked by the caspase inhibitor z-Val-Ala-Asp-fluoromethylketone either. Diphenylene ionodium (DPI), an inhibitor of NADPH oxidase, was able to prevent Tam-induced cell death but not cytochrome c release, which suggests that ROS act distal to cytochrome c. The pure antiestrogen ICI 182780 (1 microM) could partly oppose the effect of Tam in estrogen receptor positive MCF-7 cells, but not in estrogen receptor negative MDA-MB-231 cells. Pre-culturing MCF-7 cells in the absence of 17beta-estradiol (E(2)) or in the presence of a low Tam concentration (1 microM) made the cells even more susceptible to rapid death induction by 5 or 7 microM Tam. This effect was associated with decreased levels of the anti-apoptotic proteins Bcl-X(L) and Bcl-2. In conclusion, our results demonstrate induction of a rapid mitochondrial cell death program in breast cancer cells at pharmacological concentrations of Tam, which are achievable in tumor tissue of Tam-treated breast cancer patients. These mechanisms may contribute to the ability of Tam therapy to induce death of breast cancer cells.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Bongkrekic Acid/pharmacology , Breast Neoplasms/enzymology , Caspase 9/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogens/deficiency , Female , Fulvestrant , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/enzymology , Onium Compounds/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Toremifene/pharmacology
11.
Apoptosis ; 10(3): 583-95, 2005 May.
Article in English | MEDLINE | ID: mdl-15909120

ABSTRACT

Glucocorticoid-induced osteoporosis may be at least in part due to the increased apoptosis of osteocytes. To study the role of osteocyte apoptosis in glucocorticoid-induced osteoporosis, we isolated primary osteocytes from murine calvaria for the analysis of the effects of dexamethasone in in vitro culture. The cells were identified by morphology, cytochemical staining, immunocytochemical staining and mRNA expression of phosphate-regulating gene with homology to endopeptidases on the X chromosome (PHEX) and sclerosteosis/van Buchem disease gene (SOST). We found that dexamethasone induced osteocyte apoptosis in a dose-dependent manner. A glucocorticoid receptor antagonist, mifepristone (RU486), suppressed dexamethasone-induced osteocyte apoptosis, suggesting that it was mediated by glucocorticoid receptor. Immunocytochemical stainings showed that glucocorticoid receptors are present in primary osteocytes, and they were translocated to nuclei after the exposure to dexamethasone. Addition of estrogen prevented glucocorticoid receptor translocation into nuclei. Corresponding antiapoptotic effects in primary osteocytes were also seen after the pretreatment of primary osteocytes with a picomolar concentration of estrogen. The pure antiestrogen ICI 182,780 inhibited estrogen effect on apoptosis induced by dexamethasone. These data suggest that glucocorticoid receptors play an important role in glucocorticoid-induced osteocyte apoptosis. Most importantly, estrogen has a protective effect against osteocyte apoptosis. To conclude, the mechanism of glucocorticoid-induced osteoporosis may be due to the apoptosis of osteocytes, which can be opposed by estrogen.


Subject(s)
Apoptosis/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Osteocytes/drug effects , Animals , Cell Nucleus/metabolism , Cells, Cultured , Estradiol/analogs & derivatives , Estradiol/pharmacology , Fulvestrant , In Situ Nick-End Labeling , Mice , Mifepristone/pharmacology , Osteocytes/cytology , Receptors, Glucocorticoid/antagonists & inhibitors
12.
J Steroid Biochem Mol Biol ; 93(2-5): 277-83, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15860271

ABSTRACT

Experimental data suggest that sex steroids have a role in the development of breast and prostate cancers. The biological activity of sex steroid hormones in target tissues is regulated by several enzymes, including 17beta-hydroxysteroid dehydrogenases (17HSD). Changes in the expression patterns of these enzymes may significantly modulate the intracellular steroid content and play a pathophysiological role in malignant transformation. To further clarify the role of 17HSDs in breast cancer, we analyzed the mRNA expressions of the 17HSD type 1, 2, and 5 enzymes in 794 breast carcinoma specimens. Both 17HSD type 1 and 2 mRNAs were detected in normal breast tissue from premenopausal women but not in specimens from postmenopausal women. Of the breast cancer specimens, 16% showed signals for 17HSD type 1 mRNA, 25% for type 2, and 65% for type 5. No association between the 17HSD type 1, 2, and 5 expressions was detected. The patients with tumors expressing 17HSD type 1 mRNA or protein had significantly shorter overall and disease-free survival than the other patients. The expression of 17HSD type 5 was significantly higher in breast tumor specimens than in normal tissue. The group with 17HSD type 5 overexpression had a worse prognosis than the other patients. Cox multivariate analyses showed that 17HSD type 1 mRNA, tumor size, and ERalpha had independent prognostic significance. Using an LNCaP prostate cancer cell line, we developed a cell model to study the progression of prostate cancer. In this model, androgen-sensitive LNCaP cells are transformed in culture conditions into more aggressive, androgen-independent cells. The model was used to study androgen and estrogen metabolism during the transformation process. Our results indicate that substantial changes in androgen and estrogen metabolism occur in the cells during the process. A remarkable decrease in oxidative 17HSD activity was seen, whereas reductive activity seemed to increase. Since local steroid metabolism controls the bioavailability of active steroid hormones of target tissues, the variations in steroid-metabolizing enzymes during cancer progression may be crucial in the regulation of the growth and function of organs.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Cell Transformation, Neoplastic/metabolism , 17-Hydroxysteroid Dehydrogenases/classification , 17-Hydroxysteroid Dehydrogenases/genetics , Androgens/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/etiology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Estrogens/metabolism , Female , Humans , In Situ Hybridization , Male , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/etiology , Neoplasms, Hormone-Dependent/genetics , Oxidation-Reduction , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/etiology , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
13.
Calcif Tissue Int ; 75(3): 253-61, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15148559

ABSTRACT

Bone remodeling involves old bone resorption by osteoclasts and new bone formation by osteoblasts. However, the precise cellular mechanisms underlying these consecutive events remain obscure. To address this question in vitro, we have established a cell culture model in which the resorption lacunae are first created by osteoclasts and osteoblast-like cells accomplish the subsequent bone formation. We isolated osteoclasts from rat bone marrow and cultured them on bovine bone slices for 48 hours to create resorption lacunae. After removing osteoclasts, confluent differentiated primary osteoblast cultures were trypsinized and the cells were replaced on the resorbed bone slices for up to 14 days. The cultures were then examined by confocal microscopy, field emission scanning electron microscopy (FESEM), and transmission electron microscopy (TEM). Our data suggest that after osteoclastic bone resorption, osteoblast-like cells, not macrophages, remove the remaining organic matrix in the lacuna. After cleaning the lacuna, osteoblast-like cells deposit new collagen fibrils at the bottom of the lacuna and calcify the newly formed matrix only, as visualized by labeled tetracycline accumulation merely in the lacuna during the osteoblast culture. Furthermore, an electron-dense layer rich in osteopontin separates the old and new matrices suggesting formation of the cement line. Since the morphology of the newly formed matrix is similar to the natural bone with respect to the cement line and osteoid formation as well as matrix mineralization, the present method provides for the first time a powerful in vitro method to study the cellular mechanisms leading to bone remodeling also in vivo.


Subject(s)
Bone Remodeling/physiology , Bone Resorption/metabolism , Calcification, Physiologic/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Animals , Cells, Cultured , Humans , Immunohistochemistry , In Vitro Techniques , Macrophages/metabolism , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Osteoblasts/ultrastructure , Osteoclasts/ultrastructure , Osteopontin , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/metabolism
14.
Mol Cell Endocrinol ; 215(1-2): 83-8, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-15026178

ABSTRACT

17 beta-Hydroxysteroid dehydrogenases (17HSDs) regulate the biological activity of sex steroid hormones in a variety of tissues by catalyzing the interconversions between highly active steroid hormones, e.g. estradiol and testosterone, and corresponding less active hormones, estrone and androstenedione. Epidemiological and endocrine evidence indicates that estrogens play a role in the etiology of breast cancer, while androgens are involved in mechanisms controlling the growth of normal and malignant prostatic cells. Using LNCaP prostate cancer cell lines, we have developed a cell model to study the progression of prostate cancer. In the model LNCaP cells are transformed in culture condition into more aggressive cells. Our data suggest that substantial changes in androgen and estrogen metabolism occur in the cells, leading to increased production of active estrogens during the process. In breast cancer, the reductive 17HSD type 1 activity is predominant in malignant cells, while the oxidative 17HSD type 2 mainly seems to be present in non-malignant breast epithelial cells. Deprivation of an estrogen response by using specific 17HSD type 1 inhibitors is a tempting approach in treating estrogen-dependent breast cancer. Our recent studies demonstrate that in addition to sex hormone target tissues, estrogens may be important in the development of cancer in some other tissues previously not considered to be estrogen target tissues, such as the gastrointestinal tract.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Gonadal Steroid Hormones/metabolism , Neoplasms/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Oxygen/metabolism
15.
J Steroid Biochem Mol Biol ; 92(4): 281-6, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15663991

ABSTRACT

The growth and function of the prostate is dependent on androgens. The two predominant androgens are testosterone, which is formed in the testis from androstenedione and 5alpha-dihydrotestosterone, which is formed from testosterone by 5alpha-reductases and is the most active androgen in the prostate. Prostate cancer is one of the most common cancers among men and androgens are involved in controlling the growth of androgen-sensitive malignant prostatic cells. The endocrine therapy used to treat prostate cancer aims to eliminate androgenic activity from the prostatic tissue. Most prostate cancers are initially responsive to androgen withdrawal but become later refractory to the therapy and begin to grow androgen-independently. Using LNCaP prostate cancer cell line we have developed a cell model to study the progression of prostate cancer. In the model androgen-sensitive LNCaP cells are transformed in culture conditions into more aggressive, androgen-independent cells. The model was used to study androgen and estrogen metabolism during the transformation process. Our results indicate that substantial changes in androgen and estrogen metabolism occur in the cells during the process. A remarkable decrease in the oxidative 17beta-hydroxysteroid dehydrogenase activity was seen whereas the reductive activity seemed to increase. The changes suggest that during transformation estrogen influence is increasing in the cells. This is supported by the cDNA microarray screening results which showed over-expression of several genes up-regulated by estrogens in the LNCaP cells line representing progressive prostate cancer. Since local steroid metabolism controls the bioavailability of active steroid hormones in the prostate, the variations in steroid-metabolizing enzymes during cancer progression may be crucial in the regulation of the growth and function of the organ.


Subject(s)
Gonadal Steroid Hormones/metabolism , Prostatic Neoplasms/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Androgens/biosynthesis , Androgens/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Dihydrotestosterone/metabolism , Estrogens/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gonadal Steroid Hormones/biosynthesis , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Testosterone/biosynthesis , Testosterone/metabolism
17.
J Steroid Biochem Mol Biol ; 83(1-5): 119-22, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12650708

ABSTRACT

17 beta-Hydroxysteroid dehydrogenases (17HSDs) catalyze the interconversions between active 17 beta-hydroxysteroids and less-active 17-ketosteroids thereby affecting the availability of biologically active estrogens and androgens in a variety of tissues. The enzymes have different enzymatic properties and characteristic cell-specific expression patterns, suggesting differential physiological functions for the enzymes. Epidemiological and endocrine evidence indicate that estrogens play a key role in the etiology of breast cancer while androgens are involved in mechanisms controlling the growth of prostatic cells, both normal and malignant. Recently, we have developed, using LNCaP prostate cancer cell lines, a cell model to study the progression of prostate cancer. In the model LNCaP cells are transformed in culture condition to more aggressive cells, able to grow in suspension cultures. Our results suggest that substantial changes in androgen and estrogen metabolism occur in the cells during the process. These changes lead to increased production of active estrogens during transformation of the cells. Data from studies of breast cell lines and tissues suggest that the oxidative 17HSD type 2 may predominate in human non-malignant breast epithelial cells, while the reductive 17HSD type 1 activity prevails in malignant cells. Deprivation of an estrogen response by using specific 17HSD type 1 inhibitors is a tempting approach to treat estrogen-dependent breast cancer. Our recent studies demonstrate that in addition to sex hormone target tissues, estrogens may be important in the development of cancer in some other tissues previously not considered as estrogen target tissues such as colon. Our data show that the abundant expression of 17HSD type 2 present in normal colonic mucosa is significantly decreased during colon cancer development.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Neoplasms/enzymology , Breast Neoplasms/enzymology , Cell Line , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Disease Progression , Female , Humans , Male , Oxygen/metabolism , Prostatic Neoplasms/enzymology , Protein Isoforms , Tumor Cells, Cultured
18.
J Steroid Biochem Mol Biol ; 78(5): 389-400, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11738549

ABSTRACT

The Shionogi 115 (S115) mouse mammary tumor cells express the MMTV-specific 1.7 kb mRNA (orf) at a high level in the presence of androgens. In lymphoid cells the orf-gene encodes a superantigen which has an important role in establishing self-tolerance but in mammary and breast cancer cells the function of the orf gene is unclear. In the present work we studied the expression of the S115 mammary tumor cell orf sequence and its role in the androgen regulated growth of S115 cells. The cloning and sequencing of the cDNA specific for the 1.7 kb mRNA from the S115 mouse mammary tumor cells revealed a 990 bp DNA sequence with a 99.8% homology to the Mtv-17 proviral strain. There was a difference of only one amino acid (isoleu-tyr) in the coding region. A peptide was synthesized according to the hypervariable C-terminal part of the predicted protein and used to raise a rabbit antiserum. The anti-S115-orf antiserum immunoprecipitated an approximately 45 kDa protein from the metabolically labeled S115 cell lysates. In order to analyze the putative functions of the protein, the orf-sequence was linked to MoMLV-LTR and to the human ss-actin promoter in the mammalian expression vectors pLTRpoly and pHssAPr-1-neo, respectively, and transfected into NIH3T3 and S115 cells. NIH3T3 transfectants expressing orf mRNA did not show a transformed phenotype in vitro. The S115 orf transfectants proliferated somewhat more slowly than the vector transfected control cells in cell culture, both in the presence or absence of androgen, but there was no obvious change in the phenotype of S115 cells or in expression of the fibroblast growth factor 8 (FGF-8). This factor is activated by Mtv-6 integration and mediates androgen effects in these cells. Unexpectedly, however, the formation of tumors by S115 orf cells in nude mice was considerably prolonged and tumor growth retarded when compared with vector transfected control or parent S115 cells. The results suggest that MMTV-orf can be functional in breast cancer cells but the mechanism of the growth repressive effect in mammary tumor remains to be analyzed.


Subject(s)
Androgens/pharmacology , Genes, Viral , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Division/drug effects , Cloning, Molecular , DNA, Complementary/genetics , DNA, Viral/genetics , Female , Gene Expression , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/virology , Open Reading Frames , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Viral/genetics , Sequence Homology, Amino Acid , Transfection , Tumor Cells, Cultured
19.
Endocrinology ; 142(12): 5371-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713237

ABSTRACT

Decreased E2 levels after menopause cause bone loss through increased penetrative resorption. The reversal effect of E2 substitution therapy is well documented in vivo, although the detailed mechanism of action is not fully understood. To study the effects of E2 on bone resorption, we developed a novel in vitro bone resorption assay in which degradation of inorganic and organic matrix could be measured separately. E2 treatment significantly decreased the depth of resorption pits, although the area resorbed was not changed. Electron microscopy further revealed that the resorption pits were filled with nondegraded collagen, suggesting that E2 disturbed the organic matrix degradation. Two major groups of proteinases, matrix metalloproteinases (MMPs) and cysteine proteinases, have been suggested to participate in organic matrix degradation by osteoclasts. We show here that MMP-9 released a cross-linked carboxyl-terminal telopeptide of type I collagen from bone collagen, and cathepsin K released another C-terminal fragment, the C-terminal cross-linked peptide of type I collagen. E2 significantly inhibited the release of the C-terminal cross-linked peptide of type I collagen into the culture medium without affecting the release of cross-linked carboxyl-terminal telopeptide of type I collagen in osteoclast cultures. These results suggest that organic matrix degradation is initiated by MMPs and continued by cysteine proteases; the latter event is regulated by E2.


Subject(s)
Bone Matrix/metabolism , Bone Resorption/pathology , Estradiol/pharmacology , Osteoclasts/drug effects , Osteoclasts/physiology , Animals , Biomarkers , Bone and Bones/metabolism , Cathepsin K , Cathepsins/pharmacology , Cell Count , Cells, Cultured , Cellular Senescence/physiology , Collagen/metabolism , In Vitro Techniques , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Microscopy, Electron, Scanning , Osteoclasts/ultrastructure , Protease Inhibitors/pharmacology , Rats , Recombinant Proteins
20.
Lab Invest ; 81(6): 815-26, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11406643

ABSTRACT

SUMMARY: Fibroblast growth factor 8 (FGF-8) is implicated in growth of prostate cancer. Alternative splicing of the human FGF-8 gene potentially allows coding for four protein isoforms (a, b, e, and f). These isoforms differ in their binding to FGF receptors (FGFR) and in their mitogenic and transforming capacity in transfection assays. Here, we used RT-PCR and immunohistochemistry to study the expression of FGF-8 and FGFR isoforms in human prostate cancer (n = 31). Nonmalignant prostate specimens from cystoprostatectomies (n = 24) were examined as controls. Most prostate cancer samples and some control prostates also contained prostatic intraepithelial neoplasia (PIN) lesions. FGF-8a and e were expressed at significantly higher frequencies in prostate cancer (FGF-8a, 55%; FGF-8e, 45%) than in control samples (FGF-8a, 17%, p = 0.0052; FGF-8e, 8%, p = 0.0031). On the contrary, FGF-8b was found at an equal frequency in prostate cancer (55%) and in control prostates (50%). Furthermore, a combination of two or three FGF-8 isoforms (a, b, and/or e) was also expressed at a higher frequency in prostate cancer than in control samples (45% and 8%, respectively, p = 0.0031). Immunohistochemistry with an antibody recognizing all FGF-8 isoforms was more strongly immunoreactive in prostate cancer cells and PIN lesions than in normal-type epithelium. The receptor splicing variants FGFR1IIIc and FGFR2IIIc, which are activated by FGF-8, were found both in prostate cancer and control samples. Interestingly, immunoreactivity for FGFR1 and FGFR2 was much stronger in prostate cancer cells and PIN than in normal epithelium. These results demonstrate, for the first time, that FGF-8 isoforms and their receptors FGFR1IIIc and FGFR2IIIc are expressed at an increased level not only in prostate cancer but also in premalignant PIN lesions. These data suggest that FGF-8 may have an important autocrine role in the development of human prostate cancer. In addition to FGF-8b, the FGF-8 isoforms a and e may be involved in this process.


Subject(s)
Fibroblast Growth Factors/metabolism , Precancerous Conditions/metabolism , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Aged , Fibroblast Growth Factor 8 , Humans , Immunohistochemistry , Male , Middle Aged , Prostate/metabolism , Protein Isoforms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1 , Receptor, Fibroblast Growth Factor, Type 2 , Reference Values , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...