37 research outputs found

    Additive growth inhibitory effects of ibandronate and antiestrogens in estrogen receptor-positive breast cancer cell lines

    Get PDF
    INTRODUCTION: Bisphosphonates are inhibitors of osteoclast-mediated tumor-stimulated osteolysis, and they have become standard therapy for the management of bone metastases from breast cancer. These drugs can also directly induce growth inhibition and apoptosis of osteotropic cancer cells, including estrogen receptor-positive (ER+) breast cancer cells. METHODS: We examined the anti-proliferative properties of ibandronate on two ER+ breast cancer cell lines (MCF-7 and IBEP-2), and on one ER negative (ER-) cell line (MDA-MB-231). Experiments were performed in steroid-free medium to assess ER regulation and the effect of ibandronate in combination with estrogen or antiestrogens. RESULTS: Ibandronate inhibited cancer cell growth in a dose- and time-dependent manner (approximate IC(50): 10(-4 )M for MCF-7 and IBEP-2 cells; 3 × 10(-4 )M for MDA-MB-231 cells), partly through apoptosis induction. It completely abolished the mitogenic effect induced by 17β-estradiol in ER+ breast cancer cells, but affected neither ER regulation nor estrogen-induced progesterone receptor expression, as documented in MCF-7 cells. Moreover, ibandronate enhanced the growth inhibitory action of partial (4-hydroxytamoxifen) and pure (ICI 182,780, now called fluvestrant or Faslodex™) antiestrogens in estrogen-sensitive breast cancer cells. Combination analysis identified additive interactions between ibandronate and ER antagonists. CONCLUSION: These data constitute the first in vitro evidence for additive effects between ibandronate and antiestrogens, supporting their combined use for the treatment of bone metastases from breast cancer

    A Age-related inhibitory activity of rat bone marrow supernatant on osteoblast proliferation

    No full text
    Because histomorphometric indices of bone formation (osteoblastic index, tetracyclin-labeled perimeter) are deeply depressed in aged rats, while in vitro proliferation of trabecular bone cells was found increased, we hypothesized that a signal to proliferate, correctly induced by increased strains on scarce bone, could be opposed in vivo by an inhibitor present in the bone marrow extracellular medium. Thus, we tested the effect of bone marrow extracellular fluid (BM supernatant) of rat femoral diaphysis on cultures of primary osteoblasts and osteoblastic cell lines and found that it inhibited bone cell proliferation. In a group of 69 female rats aged 4, 12, and 15/21 months, there was a stepwise increase in the inhibitory activity of the BM supernatant. The double reciprocal plots relating inhibition power of the medium to BM supernatant dilution suggest that we deal with a simple system and that the kinetics of the phenomenon are the same in older and younger animals. Moreover, proliferation inhibition by BM supernatant and trabecular bone surface measured by histomorphometry in the distal femoral metaphysis were inversely correlated. Because the extracellular fluid of bone marrow is also the medium surrounding the osteoblasts and their precursor cells, our results suggest that the bone marrow negatively regulates osteogenic cells and that this inhibition could contribute to the inability of older animals to supply osteoblasts to bone in proportion to the demand. Preliminary biochemical characterization of the inhibitor suggests it to be a protein of 30-40 kDa with an isoelectric point (pI) of about 6.5.Journal ArticleSCOPUS: ar.jFLWNOinfo:eu-repo/semantics/publishe

    Determination of RNA expression for cholecystokinin/gastrin receptors (CCKA, CCKB and CCKC) in human tumors of the central and peripheral nervous system.

    No full text
    Gastrin (G17) belongs to the cholecystokinin (CCK) peptide family widely distributed in the brain, and we were the first to show that it significantly modulates the growth and migration features of tumor astyrocytes. Conflictual data have been published as to whether CCKA, CCKB and CCKC receptors are, or are not, present in tumors of the central and peripheral nervous system (CPNS) in general, and in gliomas in particular. In the present study we employed polymerase chain reaction (PCR) on a series of 29 CNPS tumors, including 20 gliomas (17 astrocytic and 3 oligodendroglial tumors), 4 schwannomas and 5 meningiomas to investigate whether RNAs were present or absent in the case of these CCKA, CCKB and CCKC receptors. The presence of the three CCK receptor subtypes was also assayed on three experimental models, i.e. the U373 human glioma, the C6 rat glioma and the 9L rat gliosarcoma. The data show that 9/20 (45%) of the gliomas exhibited RNAs for the CCKB receptor as did the C6 rat glioma, 13/20 (65%) RNAs for the CCKC receptor as did the U373 human glioma and the 9L rat gliosarcoma. Of the 20 gliomas, 17 (85%) expressed RNAs for either the CCKB or the CCKC receptor (or both), a feature which was also observed in the experimental models. One schwannoma and one meningioma exhibited RNAs for the CCKB receptor, while 4/4 schwannomas and 4/5 meningiomas showed RNAs for the CCKC receptor. None of the gliomas, schwannomas or meningiomas exhibited RNAs for the CCKA receptor, which were found in the 9L rat gliosarcoma model only. These data emphasize that 85% of the gliomas under study and 86% (25/29) of the tumors of the central and peripheral nervous system exhibited CCKB and/or CCKC receptors. This therefore suggests an important role for gastrin in the biological development of these tumors.Journal ArticleResearch Support, Non-U.S. Gov'tinfo:eu-repo/semantics/publishe

    Prolactin up-regulates cathepsin B and D expression in minor salivary glands of patients with Sjögren's syndrome.

    No full text
    Various proteases are expressed in the minor salivary glands (MSG) of patients with Sjögren's syndrome (SS), and as we have already shown, prolactin is neosynthesized in the acinar cells of patients with SS. The present study aims to characterize the influence of PRL on the expression of cathepsin B and D in the MSG of patients with SS. Cathepsin B and D expression was investigated immunohistochemically in MSG of 30 patients with SS and 15 healthy volunteers. The presence of cathepsin B and D mRNAs was checked in three SS patients and three control subjects by means of reverse transcription-polymerase chain reaction (RT-PCR). The specificity of the anti-cathepsin B and D antibodies used for the immunohistochemistry was checked by means of western blotting analysis. The influence of prolactin on the immunohistochemical expression of cathepsin B and D was quantitatively assayed by computer-assisted microscopy at three different doses (5, 50, and 500 ng/ml) on eight MSGs (four control subjects and four patients with SS) maintained ex vivo under organotypic cultures. This influence was also investigated at the mRNA level. Whereas cathepsin B immunopositivity was absent from glandular epithelial cells of healthy subjects and only slightly present in SS patients, cathepsin D immunoreactivity was considerably greater (p < 0.0001) in both the acini and the ducts of patients with SS as compared with control subjects. Cathepsin B, but not D, was also expressed in about 20% of infiltrating mononuclear cells of SS patients. Treatment of both healthy and SS minor salivary glands with PRL significantly (p < 0.05 top < 0.0001) enhanced cathepsin B and D expression in acinar and ductal cells at both protein and mRNA levels. PRL produced locally in MSGs of SS patients, but not those of healthy subjects, could play a role in the pathogenesis of Sjogren's syndrome, if only through the activation of proteolytic activity on the part of cathepsins B and D.Journal ArticleResearch Support, Non-U.S. Gov'tSCOPUS: ar.jinfo:eu-repo/semantics/publishe

    Effect of nuclear export inhibition on estrogen receptor regulation in breast cancer cells

    No full text
    We used the Crm1 inhibitor leptomycin B (LMB) to examine a possible involvement of nuclear export in estrogen receptor alpha (ER) level and function in MCF-7 breast carcinoma cells. As revealed by immunofluorescence microscopy and western blotting with anti-ER antibodies, LMB produced an accumulation of ER in cell nuclei. LMB also partly abrogated ER elimination resulting from Hsp90 disruption and 17beta-estradiol (E(2))-induced ER downregulation. By contrast, it was ineffective on ER downregulation caused by the pure anti-estrogen fulvestrant. Finally, LMB inhibited E(2)-induced progesterone receptor expression and the expression of an estrogen response element-driven luciferase reporter gene in unstimulated and E(2)-stimulated cells. Altogether, the data reported here suggest that: i) ER undergoes nuclear export directly or indirectly involving exportin Crm1; ii) degradation of unliganded and of agonist-bound ER probably occurs in an extranuclear compartment, while it is not the case for ER bound to a pure anti-estrogen; and iii) optimal ER-mediated gene transactivation seems to require nucleocytoplasmic shuttle of the receptor.SCOPUS: ar.jinfo:eu-repo/semantics/publishe

    Farnesol, a mevalonate pathway intermediate, stimulates MCF-7 breast cancer cell growth through farnesoid-X-receptor-mediated estrogen receptor activation

    No full text
    Farnesoid X receptor (FXR) is a metabolic nuclear receptor expressed in the liver and traditionally considered as a bile acid sensor. Yet, FXR has been recently demonstrated in other tissues and cells, such as the kidneys, the adrenals, and arterial smooth muscle cells. Immunohistochemical data reported in this study point to the expression of FXR in human breast cancer. In addition, FXR expression was also found by Western blotting and immunofluorescence microscopy in breast-cancer-derived cell lines MCF-7 (estrogen receptor [ER]-positive) and MDA-MB-231 (ER-negative). The FXR activator farnesol, a mevalonate pathway intermediate, exerts a mitogenic effect on MCF-7 cells. The growth stimulation is completely suppressed by antiestrogens. In contrast, MDA-MB-231 cells appear farnesol-insensitive, suggesting an involvement of ER in farnesol mitogenicity. In accordance with this interpretation, farnesol induces in MCF-7 cells a decrease of ER level, consistent with a phenomenon of receptor downregulation. Farnesol also increases progesterone receptor (PgR) expression in MCF-7 cells and stimulates ER-mediated gene transactivation in MVLN cells (MCF-7 cells stably transfected with an ER reporter gene). Of note, both effects of farnesol on ER expression and activity are completely suppressed by antiestrogens. In addition, farnesol-induced PgR is markedly reduced by FXR gene silencing (siRNA), demonstrating the involvement of FXR in the estrogenic effects of farnesol. Finally, coimmunoprecipitation experiments (FXR immunoprecipitation followed by Western blot analysis of ER in the immunoprecipitate) produced definite evidence that FXR interacts with ER. Altogether, these observations reveal the hitherto unreported presence of FXR in breast cancer and show that the latter receptor functionally interacts with ER. The occurrence of such a crosstalk calls for some caution regarding the pharmacological use of FXR agonists.SCOPUS: ar.jinfo:eu-repo/semantics/publishe
    corecore