Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
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.
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
4.
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
5.
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
6.
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
8.
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
9.
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
10.
Oncogene ; 20(22): 2791-804, 2001 May 17.
Article in English | MEDLINE | ID: mdl-11420691

ABSTRACT

Fibroblast growth factor 8 (FGF-8) is a secreted heparin-binding protein, which has transforming potential. Alternative splicing of the mouse Fgf-8 gene potentially codes for eight protein isoforms (a-h) which differ in their transforming capacity in transfected cells. S115 mouse mammary tumor cells express a transformed phenotype and secrete FGF-8 in an androgen-dependent manner. In order to study the role of FGF-8 isoforms in the induction of transformed phenotype of breast cancer cells, we over-expressed FGF-8 isoforms a, b and e in S115 cells. Over-expression of FGF-8b, but not FGF-8a or FGF-8e, induced androgen and anchorage independent growth of S115 cells. FGF-8b-transfected S115 cells formed rapidly growing tumors with increased vascularization when injected s.c. into nude mice. FGF-8a also slightly increased tumor growth and probably tumor vascularization but FGF-8e was not found to have any effects. The angiogenic activity of FGF-8b and heparin-binding growth factor fraction (HBGF) of S115 cell conditioned media was tested in in vitro and in vivo models for angiogenesis using immortomouse brain capillary endothelial cells (IBEC) and chorion allantoic membrane (CAM) assays. Recombinant FGF-8b protein was able to stimulate proliferation, migration, and vessel-like tube formation of IBECs. In addition, stimulatory effect of S115-HBGF on IBE cell proliferation was evident. A positive angiogenic response to FGF-8b was also seen in CAM assay. The results demonstrate that the expression of Fgf-8b is able to promote vessel formation. Angiogenic capacity probably markedly contributes to the ability of FGF-8b to increase tumor growth of androgen-regulated S115 mouse breast cancer cells.


Subject(s)
Cell Line, Transformed/pathology , Fibroblast Growth Factors/physiology , Mammary Neoplasms, Experimental/blood supply , Neoplasm Proteins/physiology , Neovascularization, Pathologic/etiology , Animals , Cell Adhesion , Cell Division/drug effects , Female , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Phenotype , Testosterone/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
11.
Cancer Res ; 61(10): 4229-37, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358849

ABSTRACT

Fibroblast growth factor 8 (FGF-8) is a secreted heparin-binding protein, which has mitogenic and transforming activity. Increased expression of FGF-8 has been found in human breast cancer, and it has a potential autocrine role in its progression. Human FGF-8 is alternatively spliced to generate four protein isoforms (a, b, e, and f). Isoform b has been shown to be the most transforming. In this work, we studied the role of FGF-8b in the growth (in vitro and in vivo) of MCF-7 human breast cancer cells, which proliferate in an estrogen-dependent manner. Constitutive overexpression of FGF-8b in MCF-7 cells down-regulated FGF-8b-binding receptors FGF receptor (FGFR) 1IIIc, FGFR2IIIc, and FGFR4 found to be expressed in these cells. FGF-8b overexpression led to an increase in the anchorage-independent proliferation rate in suspension culture and colony formation in soft agar, when MCF-7 cells were cultured with or without estradiol. FGF-8b also provided an additional growth advantage for cells stimulated with estradiol. In addition, FGF-8b-transfected cells invaded more actively through Matrigel than did control cells. This was possibly due to the increased secretion of matrix metalloproteinase 9. In vivo, FGF-8b-transfected MCF-7 cells formed faster growing tumors than vector-only-transfected cells when xenografted into nude mice. The tumors formed by FGF-8b-transfected cells were more vascular than the tumors formed by vector-only-transfected cells. In conclusion, FGF-8b expression confers a growth advantage to MCF-7 breast carcinoma cells, both in vitro and in vivo. In addition to stimulation of proliferation, this growth advantage probably arises from increased invasion and tumor vascularization induced by FGF-8b. The results suggest that FGF-8b signaling may be an important factor in the regulation of tumorigenesis and progression of human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fibroblast Growth Factors/biosynthesis , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Cell Adhesion/physiology , Cell Division/drug effects , Cell Division/physiology , Female , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neovascularization, Pathologic/metabolism , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/pharmacology , Receptors, Fibroblast Growth Factor/biosynthesis , Receptors, Fibroblast Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous
13.
Endocrinology ; 140(11): 5412-21, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537173

ABSTRACT

PRL is one of several polypeptide factors that regulate growth and differentiation of prostate epithelium besides steroid hormones. This hormone may also participate in the development of pathologic changes of the prostate, as evidenced by marked prostate hyperplasia in hyperprolactinemic mice. We have previously demonstrated expression of PRL receptors and androgen-dependent local production of PRL in rat and human prostate epithelium, suggesting the existence of an autocrine loop. We now show that PRL acts as a survival factor for epithelial cells of rat dorsal and lateral prostate but not ventral prostate, using long-term organ cultures as an in vitro model. Culture of prostate explants in androgen-free medium was associated with a transient surge of apoptosis during the first 2-4 days of culture in rat ventral, dorsal, and lateral prostate tissues, as quantified by either nuclear morphology or in situ DNA fragmentation analysis. PRL significantly inhibited apoptosis in androgen-deprived dorsal and lateral prostate cultures, by 40-60%, as determined by the two methods. The present study has established conditions and methodology for analysis of apoptosis in organ cultures of rat prostate and suggests a physiological role for PRL as a survival factor for prostate epithelium.


Subject(s)
Androgens/administration & dosage , Apoptosis/drug effects , Organ Culture Techniques , Prolactin/pharmacology , Prostate/cytology , Animals , Culture Media , DNA Fragmentation , Epithelial Cells/cytology , Male , Mitosis , Rats , Rats, Sprague-Dawley
14.
Bone ; 25(2): 211-5, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10456387

ABSTRACT

The increase of bone resorption and reduction of bone mass in postmenopausal women can be prevented by treatment with estrogen. Although it is well established that estrogen treatment normalizes the increased bone turnover, the mechanism by which estrogen exerts its protective influence at the cellular and molecular level in bone remains elusive. It has been shown that osteoblasts are involved in osteoclast development and osteoclastic bone resorption. In this work we examine the effect of estrogen (E2) on osteoclast-mediated bone resorption via the medium conditioned by osteoblast cultures. The conditioned medium collected from osteoblast cultures without (CM) or with 0.1 nmol/L 17beta-estradiol (E-CM) was mixed in a 1:1 ratio with fresh osteoclast culture medium. Osteoclasts were isolated from the bone marrow of 3-day-old NMRI mice and cultured on bovine bone slices. The total number of multinucleated tartrate-resistant alkaline phosphatase (TRAP)-positive cells in cultures with CM and E-CM was similar to that of cells incubated in control medium. However, the number of osteoclasts containing more than three nuclei was significantly smaller in the cultures containing E-CM. The total area of resorption was only slightly decreased in cultures containing CM, but was markedly inhibited in cultures with E-CM. In osteoblast cultures, the production of interleukin (IL)-1 and IL-6, but not of TNF-alpha, was reduced by 0.1 nmol/L E2. Our data suggest that E2 treatment of osteoblasts decreases the production of factor(s) that induces osteoclast differentiation to multinucleated cells with a higher capacity for bone resorption.


Subject(s)
Culture Media, Conditioned/pharmacology , Estradiol/pharmacology , Osteoblasts/drug effects , Osteoclasts/drug effects , Acid Phosphatase , Animals , Animals, Newborn , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cattle , Cells, Cultured , Female , Interleukin-1/metabolism , Interleukin-6/metabolism , Isoenzymes , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/metabolism , Tartrate-Resistant Acid Phosphatase , Tumor Necrosis Factor-alpha/metabolism
15.
Mol Cell Endocrinol ; 149(1-2): 29-40, 1999 Mar 25.
Article in English | MEDLINE | ID: mdl-10375015

ABSTRACT

Vascular endothelial growth factor (VEGF) is a major inducer of tumor angiogenesis and an important prognostic factor in breast cancer. Hypoxia is an important inducer of VEGF expression but less is known of the role of hormones in VEGF regulation. We have studied the regulation of VEGF, VEGF-B, VEGF-C, and VEGF-D mRNAs in human MCF-7 and mouse S115 breast carcinoma cells stimulated by estrogens and androgens, respectively. VEGF, VEGF-B, and VEGF-C were expressed in both cell lines, whereas VEGF-D was expressed only in S115 cells. Addition of estradiol (E2) caused a biphasic increase of VEGF mRNA in MCF-7 cells and led to accumulation of the VEGF protein in the culture medium. The VEGF-B mRNA was not affected, while a decrease occurred in VEGF-C mRNA. Similarly, testosterone upregulated the expression of VEGF mRNA in the S115 cells. Experiments with actinomycin D and cycloheximide suggested that estrogen induction of VEGF mRNA is dependent on the synthesis of new mRNA and increased mRNA half-life. The antiestrogen ICI 182.780 inhibited E2 stimulation of VEGF, suggesting that the effect was mediated by the estrogen receptor. In contrast, the antiestrogens tamoxifen and toremifene which inhibit MCF-7 cell growth in vivo and in vitro did not inhibit estrogen effect but induced VEGF mRNA expression when used alone. The antiandrogen cyprosterone acetate inhibited T induction of VEGF mRNA in S115 cells, thus suggesting that activation of androgen receptor must be involved in the increase of VEGF mRNA. Our results suggest that both estrogen and androgen stimulate the expression of VEGF by increasing gene transcription and mRNA stability. In addition, the antiestrogens tamoxifen and toremifene also increased VEGF expression. Estrogen and androgen induction of VEGF expression and promotion of new vessel formation may be an important paracrine mechanism by which these hormones contribute to the early phase of tumor growth of hormonal cancer.


Subject(s)
Breast Neoplasms/metabolism , Endothelial Growth Factors/metabolism , Estrogen Antagonists/pharmacology , Lymphokines/metabolism , Neoplasms, Hormone-Dependent/metabolism , Steroids/pharmacology , Animals , Breast Neoplasms/genetics , Cycloheximide/pharmacology , Cyproterone Acetate/pharmacology , Endothelial Growth Factors/genetics , Estradiol/pharmacology , Female , Humans , Lymphokines/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Neoplasms, Hormone-Dependent/genetics , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tamoxifen/pharmacology , Testosterone/pharmacology , Toremifene/pharmacology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
FASEB J ; 13(3): 513-22, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10064618

ABSTRACT

Carbonic anhydrase III (CA III; EC 4.2.1.1) is a cytoplasmic enzyme that exhibits a relatively low carbon dioxide hydratase activity. It is expressed at a very high level in skeletal muscle, where physical exercise has been shown to increase free radical production. In this work we show the effect of overexpression of CA III on cellular response to oxidative stress. Rat CA III cDNA was transfected to NIH/3T3 cells, which have no endogenous CA III expression. The isolated clones expressed CA III mRNA and protein. The protein was localized to cytoplasm and nuclei. Compared to parental cells, transfected cells showed lower basal oxidized state as judged by measurement of intracellular reactive oxygen species (ROS) using fluorescent dye and an image analysis system. Addition of exogenous H2O2 to cells induced a rapid increase of ROS in control but not in CA III overexpressing cells. Association of this phenomenon with CA III expression was further confirmed by showing that overexpression of CA II could not prevent H2O2-stimulated increase of ROS. In proliferation assays, CA III overexpressing cells grew faster and were more resistant to cytotoxic concentrations of H2O2 than control cells. After a 16 h exposure to oxidative stress, the number of apoptotic cells was also reduced in transfectants. Our results suggest that CA III functions as an oxyradical scavenger and thus protects cells from oxidative damage. A lower level of free radicals in CA III overexpressing cells may also affect growth signaling pathways.


Subject(s)
Apoptosis/drug effects , Carbonic Anhydrases/metabolism , Hydrogen Peroxide/pharmacology , Oxidative Stress , 3T3 Cells , Animals , COS Cells , Carbonic Anhydrases/genetics , Mice , Rats , Reactive Oxygen Species/metabolism , Transfection
17.
J Cell Biochem ; 73(4): 500-7, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10733344

ABSTRACT

Estrogens as well as some antiestrogens have been shown to prevent bone loss in postmenopausal women. These compounds seem to inhibit bone resorption, but their anabolic effects have been less explored. In this study, bone marrow cultures were used to compare the effect of 17beta-estradiol (E2), and two triphenylethylene derivatives, tamoxifen (TAM), and FC1271a, and a benzothiophene derivative raloxifene (RAL) on differentiation of osteoblasts. All enhanced osteoblastic differentiation of 21-day cultures as indicated by increased mineralization and bone nodule formation. All, except RAL, stimulated cell proliferation during the first 6 days of the culture. However, in the presence of RAL the content of total protein was increased in 13-day cultures. SDS-PAGE and autoradiography of [14C]-proline labeled proteins revealed elevated level of the newly synthesized collagen type I. The pure antiestrogen ICI 182,780 abolished the increase of the specific activity of alkaline phosphatase by E2, TAM, and FC1271a but not the effect of RAL on protein synthesis. Our results show that E2 as well as TAM, FC1271a, and RAL stimulate bone formation in vitro but the mechanism of the anabolic action of RAL in bone clearly differs from that of E2, TAM, and FC1271a.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Animals , Bone Marrow Cells/physiology , Bone Matrix/drug effects , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Female , Mice , Osteoblasts/physiology
18.
Biochem Biophys Res Commun ; 250(3): 805-8, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9784427

ABSTRACT

Fgf8 is an embryonally expressed mitogenic fibroblast growth factor which has transforming capacity. It is expressed in S115 mouse mammary tumor cells (S115 cells) and in parental tumors of DD/Sio mice as well as in some human breast and prostate cancer cell lines. In S115 cells androgens induce the expression of Fgf8 which seems to be associated with the androgen-maintained malignant phenotype of the cells. S115 cells also contain and express Mtv proviruses known to insertionally activate oncogenes in other tumor cells. Here we studied the possibility of insertional activation of Fgf8 in S115 cells by MMTV proviral integration. We demonstrate by Southern blotting that the genomic DNA from DD/Sio tumors and S115 cells contains Mtv-sequences (Mtv-6 and Mtv-17) which are not found in the DNA from spleen or liver of the DD/Sio mice. In addition, the newly integrated Mtv-6 was localized to the DNA fragment containing the Fgf8 gene. Furthermore, the expression of Fgf8 mRNA in DD/Sio tumors and S115 cells was not found in mammary gland or spleen and liver of DD/Sio mice. In S115 cells, Fgf8 mRNA expression was induced in parallel to MMTV mRNA by androgen and glucocorticoids which supports the possibility that Fgf8 is controlled by the steroid-regulated MMTV-LTR. In conclusion, our data provide evidence that the insertion of MMTV into the DD/Sio tumor DNA is associated with the transcriptional activation of Fgf8 in DD/Sio tumor and consequently in S115 mouse mammary tumor cells.


Subject(s)
Fibroblast Growth Factors/biosynthesis , Gene Expression Regulation, Neoplastic , Mammary Tumor Virus, Mouse/physiology , Virus Integration , Animals , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Viral , Genome, Viral , Humans , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics
19.
FASEB J ; 11(14): 1297-307, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9409549

ABSTRACT

Peptide hormones and growth factors are involved in the regulation of prostatic cell proliferation, differentiation, and programmed cell death, which functions are primarily controlled by androgen. In carcinogenesis, prostatic cancer cells often lose androgen dependence and become largely dependent on local growth factors. The prostatic cancer cells able to respond to factors other than androgen by proliferation and inhibition of apoptosis are possibly able to survive. We demonstrate that prostatic epithelium expresses prolactin mRNA and protein in a characteristic manner. By using in situ hybridization, an overall distribution of prolactin mRNA was demonstrated in the epithelium of rat dorsal and lateral prostate, whereas a very specific localization of prolactin protein to single cells was observed by immunohistochemistry in the same tissues. In these cells, immunoelectron microscopy showed that prolactin was primarily localized to the secretory granules. These data demonstrate a selective regulation of prostatic prolactin at least at the level of transcript processing/translation and/or protein accumulation and secretion. In addition, the expression of prolactin protein in rat dorsal and lateral prostate was found to be androgen dependent in vivo in castrated and in castrated, testosterone-treated rats, as well as in vitro in organ cultures. Our results support the concept of an autocrine/paracrine loop of prolactin action in prostate where it could mediate some of androgen actions. Also, locally synthesized prolactin might belong to the factors that take over androgen regulation of prostatic cancer cells during the development of androgen-independent growth.


Subject(s)
Androgens/metabolism , Prolactin/metabolism , Prostate/metabolism , Animals , Cytoplasmic Granules/metabolism , Epithelium/drug effects , Epithelium/metabolism , Epithelium/ultrastructure , Gene Expression Regulation/drug effects , In Situ Hybridization , Male , Microscopy, Immunoelectron , Orchiectomy , Organ Culture Techniques , Prolactin/genetics , Prostate/drug effects , Prostate/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Testis/physiology , Testosterone/pharmacology
20.
J Clin Invest ; 99(4): 618-27, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9045863

ABSTRACT

Prolactin is widely expressed in different tissues, and it is presumed to have both local and systemic actions. In males it is known to influence reproductive functions but the significance and mechanisms of prolactin action in male accessory reproductive tissues are poorly understood. Here we show that prolactin acts as a direct growth and differentiation factor for human prostate, as measured by changes in DNA synthesis and epithelial morphology of organ cultures. Furthermore, we report the expression in human prostate of a short prolactin receptor form in addition to the long form, based upon ligand cross-linking studies and RT-PCR analysis of mRNA expression. The highest density of prolactin receptors was detected in the secretory epithelial cells by immunohistochemistry. Finally, we report that prolactin is locally produced in human prostate epithelium, as evidenced by marked prolactin immunoreactivity in a significant portion of prostate epithelial cells, with parallel expression of prolactin mRNA in human prostate. Collectively, these data provide significant support for the existence of an autocrine/paracrine loop of prolactin in the human prostate and may shed new light on the involvement of prolactin in the etiology and progression of neoplastic growth of the prostate.


Subject(s)
Prolactin/biosynthesis , Prolactin/physiology , Prostate/metabolism , Receptors, Prolactin/biosynthesis , Receptors, Prolactin/physiology , Adult , Aged , DNA/metabolism , Epithelium/metabolism , Humans , Male , Middle Aged , Molecular Weight , Organ Culture Techniques , Prostate/drug effects , RNA, Messenger/biosynthesis , Receptors, Prolactin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...