Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Oncogene ; 32(19): 2390-402, 2013 May 09.
Article in English | MEDLINE | ID: mdl-22751110

ABSTRACT

Two thirds of breast cancers express estrogen receptors (ER). ER alpha (ERα) mediates breast cancer cell proliferation, and expression of ERα is the standard choice to indicate adjuvant endocrine therapy. ERbeta (ERß) inhibits growth in vitro; its effects in vivo have been incompletely investigated and its role in breast cancer and potential as alternative target in endocrine therapy needs further study. In this work, mammary epithelial (EpH4 and HC11) and breast cancer (MC4-L2) cells with endogenous ERα and ERß expression and T47-D human breast cancer cells with recombinant ERß (T47-DERß) were used to explore effects exerted in vitro and in vivo by the ERß agonists 2,3-bis (4-hydroxy-phenyl)-propionitrile (DPN) and 7-bromo-2-(4-hydroxyphenyl)-1,3-benzoxazol-5-ol (WAY). In vivo, ERß agonists induced mammary gland hyperplasia and MC4-L2 tumour growth to a similar extent as the ERα agonist 4,4',4''-(4-propyl-(1H)-pyrazole-1,3,5-triyl) trisphenol (PPT) or 17ß-estradiol (E2) and correlated with higher number of mitotic and lower number of apoptotic features. In vitro, in MC4-L2, EpH4 or HC11 cells incubated under basal conditions, ERß agonists induced apoptosis measured as upregulation of p53 and apoptosis-inducible factor protein levels and increased caspase 3 activity, whereas PPT and E2 stimulated proliferation. However, when extracellular signal-regulated kinase 1 and 2 (ERK ½) were activated by co-incubation with basement membrane extract or epidermal growth factor, induction of apoptosis by ERß agonists was repressed and DPN induced proliferation in a similar way as E2 or PPT. In a context of active ERK ½, phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/RAC-alpha serine/threonine-protein kinase (AKT) signalling was necessary to allow proliferation stimulated by ER agonists. Inhibition of MEK ½ with UO126 completely restored ERß growth-inhibitory effects, whereas inhibition of PI3K by LY294002 inhibited ERß-induced proliferation. These results show that the cellular context modulates ERß growth-inhibitory effects and should be taken into consideration upon assessment of ERß as target for endocrine treatment.


Subject(s)
Breast Neoplasms/pathology , Estrogen Receptor beta/metabolism , MAP Kinase Signaling System/physiology , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Mammary Neoplasms, Experimental/pathology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Apoptosis/physiology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Female , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction
2.
Br J Pharmacol ; 155(4): 494-504, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18604234

ABSTRACT

BACKGROUND AND PURPOSE: Breast cancer, the most common cancer in women in most countries, is a highly stressful disease. Catecholamines released during stress bind to adrenoceptors and we have recently described alpha(2)-adrenoceptors in human breast cell lines, linked to enhanced cell proliferation. The purpose was to assess the in vivo effects of compounds acting on alpha(2)-adrenoceptors in a reliable model of breast cancer. EXPERIMENTAL APPROACH: The expression of alpha(2)-adrenoceptors was confirmed by immunocytochemistry, immunofluorescence and reverse transcription-PCR in the mouse mammary tumour cell line MC4-L5. Proliferation was assessed by [(3)H]thymidine incorporation and tumours were measured daily. Apoptosis was assessed by terminal deoxynucleotidyl transferase-mediated dUTP digoxigenin nick-end labelling. KEY RESULTS: Incubation for 2 days with alpha(2)-adrenoceptor agonists (clonidine and dexmedetomidine) significantly enhanced proliferation of the mouse mammary tumour cell line MC4-L5. These agonists also significantly stimulated tumour growth of the progestin-dependent tumour C4-HD even in the presence of medroxyprogesterone acetate (MPA). In every tumour tested (C4-HD, CC4-2-HD and CC4-3-HI), regardless of MPA sensitivity, clonidine significantly enhanced tumour growth in the absence of MPA. The alpha(2)-adrenoceptor antagonists, yohimbine and rauwolscine, completely reversed the effects of clonidine. However, the group receiving yohimbine alone showed a nonsignificant but constant increase in tumour growth, whereas rauwolscine alone diminished tumour growth significantly, behaving as a reverse agonist. In CC4-3-HI tumours, rauwolscine treatment enhanced apoptosis and diminished the mitotic index, whereas clonidine had the inverse effect. CONCLUSIONS AND IMPLICATIONS: Alpha(2)-adrenoceptor agonists enhanced tumour growth and rauwolscine behaved in vivo as a reverse agonist, suggesting that it may be tested for adjuvant treatment.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Cell Proliferation/drug effects , Mammary Neoplasms, Experimental/drug therapy , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Animals , Cell Line, Tumor , Clonidine/pharmacology , Dexmedetomidine/pharmacology , Drug Inverse Agonism , Female , Mammary Neoplasms, Experimental/physiopathology , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Receptors, Adrenergic, alpha-2/metabolism , Yohimbine/pharmacology
3.
Immunology ; 104(1): 80-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11576224

ABSTRACT

In the present study we examine the effects of medroxyprogesterone acetate (MPA) on the specific antibody secretion to T-dependent antigens. Our results show that the in vivo administration of MPA to mice, 7 or 90 days before immunization with sheep red blood cells (SRBC), significantly enhanced both, primary and secondary antibody responses, without affecting delayed-type hypersensitivity (DTH). These effects could be counteracted by the anti-progestin onapristone or ZK 98299 (ZK) suggesting that MPA interacted with progesterone (PRG) receptors to increase B-cell response. To better understand the mechanisms involved in MPA activity we carried out cultures of splenocytes, bone marrow cells or lymph node cells from immunized mice in the presence of MPA, and evaluated the amount of antibody release to supernatants. We found that low doses of MPA (10(-9) M and 10(-10) M) significantly enhanced the in vitro production of specific immunoglobulin G (IgG) antibodies, an effect that appears to involve the interaction of the progestin with PRG receptors, as judged by the inhibition of MPA effects with ZK (10(-8) M) or RU486 (10(-9) M). These receptors were detected by flow cytometry analysis in a proportion of T lymphocytes. Because MPA did not increase the number of immunoglobulin-secreting cells, our findings suggest that MPA enhanced the capacity of individual cells to produce specific immunoglobulin.


Subject(s)
Immunoglobulins/biosynthesis , Medroxyprogesterone Acetate/immunology , Progesterone Congeners/immunology , Animals , Cell Culture Techniques , Dose-Response Relationship, Immunologic , Erythrocytes/immunology , Female , Hypersensitivity, Delayed/immunology , Immunologic Memory , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Receptors, Progesterone/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
4.
Cancer Res ; 61(1): 293-302, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196177

ABSTRACT

We have developed an experimental model of mammary carcinogenesis in which the administration of medroxyprogesterone acetate (MPA) to female BALB/c mice induces progestin-dependent ductal metastatic mammary tumors with high levels of estrogen receptor (ER) and progesterone receptor (PR). Through selective transplants in untreated mice, we have obtained progestin-independent variants, still expressing high levels of ER and PR. Primary cultures of the MPA-induced carcinomas C4-HD and C7-HI were set up, and after 3-4 months, several different cell lines were obtained. Four of these, MC4-L1, MC4-L2, MC4-L3, and MC4-L5 were established from C4-HD and a fifth, MC7-L1, from C7-HI. All cells were of epithelial origin, as demonstrated by electron microscopy and by immunocytochemical identification of cytokeratin and cadherin. In vitro MC4-L1, MC4-L3, and MC4-L5 showed a typical epithelial morphology; when transplanted in vivo, they originated metastatic carcinomas with different degrees of differentiation. MC4-L2 and MC7-L1 deviated from the standard epithelial picture; they disclosed a spindle-shaped morphology in vitro and in vivo gave rise to a biphasic spindle cell/tubular carcinoma and an anaplastic carcinoma, respectively; both lines gave rise to metastases. This differential morphology correlated with a higher degree of aggressiveness, as compared with MC4-L1, MC4-L3, and MC4-L5. ERs and PRs were detected by binding, immunocytochemistry, and Western blot. In vitro, MC4-L2 and MC7-L1 were stimulated by MPA (nM to microM) and 17beta-estradiol (nM and 10 nM); no significant stimulation was observed in MC4-L1, MC4-L3, and MC4-L5 under the same experimental conditions. In vivo, MPA significantly stimulated tumor growth in all epithelioid lines but not in MC4-L2 and MC7-L1. A progestin-dependent growth pattern was confirmed for MC4-L1, MC4-L3, and MC4-L5 in successive transplants, whereas MC4-L2 and MC7-L1 behaved as progestin independent. This is the first description of mouse mammary carcinoma cell lines expressing ER and PR. The different in vitro hormone responses as compared with in vivo and the differential effects of 17beta-estradiol in the parental tumors and in cell lines render these lines useful tools for the in vitro and in vivo study of hormone regulation of tumor growth and metastases.


Subject(s)
Carcinoma, Ductal, Breast/pathology , Mammary Neoplasms, Experimental/pathology , Neoplasms, Hormone-Dependent/pathology , Tumor Cells, Cultured , Animals , Carcinoma, Ductal, Breast/metabolism , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Disease Models, Animal , Estradiol/pharmacology , Female , Immunohistochemistry , Mammary Neoplasms, Experimental/metabolism , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Microscopy, Electron , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
5.
Leuk Res ; 25(2): 165-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11166832

ABSTRACT

In a previous paper we reported the occurrence of a high incidence of lymphomas in N-methyl-N-nitrosourea (MNU)-treated mice, in the course of an experiment of combined chemical-hormonal carcinogenesis in mammary gland, in which we used medroxyprogesterone acetate (MPA) and MNU in different treatment protocols. In this report we have analyzed the action of MPA in the leukemogenic effects of MNU, by specifically selecting for the analysis experimental groups in which only few mammary carcinomas had developed. A high incidence of lymphomas (65%, median latency: 176 days) was registered in MNU-treated mice, and the administration of MPA was associated with a significant reduction in the incidence of lymphomas (P<0.001) in all protocols.


Subject(s)
Lymphoma/prevention & control , Medroxyprogesterone Acetate/pharmacology , Methylnitrosourea/toxicity , Animals , Female , Immunohistochemistry , Incidence , Lymphoma/chemically induced , Mice , Mice, Inbred BALB C
6.
Cancer Detect Prev ; 23(5): 375-86, 1999.
Article in English | MEDLINE | ID: mdl-10468889

ABSTRACT

Transforming growth factor-beta2 (TGF-beta2) and -beta3 mRNA expressions were studied in ductal hormone-dependent (HD) and -independent (HI) in vivo lines of the medroxyprogesterone acetate (MPA)-induced mammary tumor model in Balb/c mice. MPA treatment of HD tumors induced a significant decrease in TGF-beta2 and -beta3 mRNA levels. Progression to an HI phenotype of ductal tumors was associated with reduced TGF-beta2 and -beta3 expressions, as compared with their HD counterparts. Exogenously added TGF-beta1, -beta2, and -beta3 (1 ng/ml) inhibited the proliferation of primary cultures of epithelial cells from ductal HD and HI tumors. In addition, TGF-beta expression and effects were studied in the other type of MPA-induced mammary tumors, which are of lobular origin and lack steroid hormone receptors and evidence an HI behavior. These lobular HI lines showed TGF-beta2 levels similar to those found in HD lines growing in MPA-treated mice. In contrast, TGF-beta3 mRNA levels were 12- to 20-fold higher than in HD tumors. Primary cultures of lobular HI epithelial cells required either TGF-beta concentrations of 10 ng/ml to show an inhibitory response, or were completely resistant to TGF-beta inhibition. Studies of the molecular mechanisms involved in reduction or loss of TGF-beta responsiveness in lobular HI tumors showed that cell surface type II TGF-beta receptor levels were lower in these tumors than those present in HD tumors. Our results support the hypothesis that TGF-beta could play a role as an autocrine growth inhibitor in HD and HI ductal tumors. Autonomous growth of lobular HI tumors could be favored by undetectable or low TGF-beta1 and -beta2 expressions and by reduced or lost sensitivity of epithelial cells to TGF-beta's antiproliferative effects. However, the extremely high levels of TGF-beta3 expression in lobular HI tumors, in spite of reduced sensitivity to TGF-beta3 inhibitory growth effect in tumor epithelial cells, suggest a net positive role for TGF-beta3 in these tumors.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Neoplasms, Hormone-Dependent/metabolism , Protein Isoforms , Transforming Growth Factor beta/pharmacology , Affinity Labels/metabolism , Animals , Blotting, Northern , Cyclin D1/metabolism , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Tumor Cells, Cultured
7.
Breast Cancer Res Treat ; 54(2): 93-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10424399

ABSTRACT

From mouse mammary progestin-dependent (PD) adenocarcinomas induced with medroxyprogesterone acetate (MPA) we developed several in vivo lines that are maintained by subcutaneous syngeneic passages in MPA-treated mice and express estrogen (ER) and progesterone receptors (PR). Although most lines remained PD, with time some progestin-independent (PI) variants arose. Both PD and PI tumors regress with estrogen treatment although estrogen-resistant variants may also arise. The object of this paper was to investigate the reversibility of estrogen-resistance and its possible relation with hormone receptor down-regulation. Tumor regression was induced in a progestin-independent tumor line (BET) by treatment with a 5 mg 17beta-estradiol (E2) silastic pellet. One of the tumors started to grow disclosing an estrogen-resistant pattern of growth. This tumor line (BET-R) was transplanted into E2-treated and untreated animals (n = 4-6), selecting for the next passage tumors growing in treated animals. Seven new sublines were obtained at different passages by selecting for the next passage the tumors that had grown in untreated mice (BET-Ra-BET-Rg), until no tumors grew in E2-treated mice. ER and PR were measured by a ligand-binding, dextran-coated charcoal method using a single saturating point. From the seven sublines initiated, the first four proved to be reversible after 3-6 generation transplantation and the last three did not revert. A difference in PR expression between BET and BET-R (p < 0.05) was registered, but it did not correlate with the specific hormone behavior since two reverted lines had a pattern similar to that of BET and the other two were similar to BET-R. The expression of PR was higher in E2-treated mice (p < 0.05) and highly variable in the parental line. This led us to study the expression of PR at different stages of the estrous cycle. Higher levels of PR were observed in proestrous, estrous, and metestrous (p < 0.05) than in diestrous, and undetectable levels were found in ovariectomized mice. No differences in ER expression were detected during the estrous cycle. It can be concluded that under certain experimental conditions, estrogen-resistance is a reversible phenomenon. The experimental manipulation of hormone resistance may help develop strategies to modify the response to anti-hormones in humans.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Hormonal/therapeutic use , Estradiol/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Animals , Cell Division , Drug Implants , Drug Resistance, Neoplasm , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate , Mice , Mice, Inbred BALB C , Progestins/toxicity , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Receptors, Progesterone/drug effects , Receptors, Progesterone/metabolism
8.
J Steroid Biochem Mol Biol ; 68(1-2): 11-21, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10215033

ABSTRACT

We have developed a model of hormonal carcinogenesis in BALB/c female mice, in which MPA induced ductal mammary adenocarcinomas, expressing high levels of estrogen and progesterone receptors (ER and PR). A series of tumor lines, retaining both PR and ER expression, were obtained from selected tumors, which are maintained by syngeneic passages. In this model progesterone behaves as the growth-stimulating hormone (progesterone-dependent or PD tumors), whereas estrogens induce tumor regression. Through selective treatments we were able to derive a series of progesterone-independent (PI) variants. These lines do not require progesterone treatment to grow in ovariectomized female BALB/c mice, but retain, however, the expression of ER and PR. The aim of this paper is to investigate a possible regulatory role of the progesterone receptor (PR) on PI tumor growth. ER and PR were detected by immunocytochemistry in all lines studied. They were also characterized using biochemical assays and Scatchard plots. No differences in Kd of PR or ER were detected in PI variants. AR or GR were not detected in tumor samples using biochemical assays. Estradiol (5 mg silastic pellet) induced complete tumor regression in all tumors tested. We also evaluated the effects of different antiprogestins on tumor growth. Onapristone (10 mg/kg/day) and mifepristone (4.5 mg/kg/day) were able to induce complete tumor regression. The antiandrogen flutamide (5 mg silastic pellet) had no effect on tumor growth in agreement with the lack of androgen receptors. We used an in vitro approach to corroborate that the antiprogestin-induced inhibition was not attributable to an intrinsic effect. Cultures of a selected PI line were treated with PR antisense oligodeoxynucleotides (ASPR) to inhibit in vitro cell proliferation. A significant decrease of 3H-thymidine uptake was observed in cells of a PI line growing in the presence of 2.5% charcoalized fetal calf serum and 0.8-20 microg/ml ASPR. It can be concluded that the PR pathway is an essential path in the growth stimulation of PI tumors.


Subject(s)
Adenocarcinoma/metabolism , Mammary Neoplasms, Experimental/metabolism , Receptors, Progesterone/metabolism , Adenocarcinoma/chemically induced , Adenocarcinoma/drug therapy , Androgens/metabolism , Animals , Binding Sites , Estradiol/pharmacology , Female , Flutamide/pharmacology , Glucocorticoids/metabolism , Gonanes/pharmacology , Hormone Antagonists/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Neoplasms, Hormone-Dependent/chemically induced , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Oligonucleotides, Antisense/pharmacology , Ovariectomy , Receptors, Estrogen/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Thymidine/metabolism , Tumor Cells, Cultured
9.
J Steroid Biochem Mol Biol ; 70(4-6): 133-42, 1999.
Article in English | MEDLINE | ID: mdl-10622401

ABSTRACT

Primary cultures of the medroxyprogesterone acetate-induced mouse mammary tumor line C4-HD are stimulated by medroxyprogesterone acetate (MPA) or progesterone. Serum obtained from ovariectomized, MPA-treated animals (OVX-MPA) exerts a stimulatory effect that is significantly higher than that induced by serum obtained from OVX mice with the exogenous addition of MPA, suggesting the involvement of MPA-induced serum factors potentiating the proliferative effect of MPA. The object of this paper is to further explore the stimulatory effect of mouse serum and to investigate the role of aFGF and bFGF on cell proliferation. The role of PR as possible mediators was tested using two different antiprogestins and antisense oligodeoxynucleotides of PR A isoform. Serum was obtained from OVX untreated or MPA-treated mice and was charcoalized and/or heat-inactivated. The effect of MPA or mifepristone at 10 nM concentrations was tested. Charcoalization and heat inactivation exerted a stimulatory effect (P<0.01) when OVX-serum was used. This effect was potentiated by MPA. Charcoalized OVX-MPA serum induced a significant inhibition of cell proliferation that was restored by the exogenous addition of MPA or by heat inactivation. Mifepristone induced an inhibition of 3H-thymidine uptake when OVX-MPA serum was used. These results suggest that serum factors activated by different manipulations may replace the stimulatory effect of MPA. When charcoalized fetal calf serum (chFCS) was used, a higher proliferative activity was obtained using higher serum concentrations. Mifepristone and onapristone 10 nM also inhibited this effect. aFGF and bFGF 100 ng/ml were both able to stimulate 3H-thymidine uptake. MPA exerted an additive effect. Mifepristone 10 nM inhibited bFGF and MPA+bFGF induced cell proliferation. Antisense oligodeoxynucleotides of PR (ASPR) were used to further confirm the participation of PR in the proliferative pathway of these cells. They inhibited serum and bFGF-induced cell proliferation in a specific dose-dependent manner. Our results suggest that PR play a central role in proliferation and suggest the existence of a cross-talk between steroid and growth factor signaling pathways.


Subject(s)
Cell Division/drug effects , Growth Substances/pharmacology , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/pharmacology , Progesterone/pharmacology , Animals , Blood , Culture Media , DNA Replication/drug effects , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Ovariectomy , Receptors, Progesterone/genetics , Thymidine/metabolism , Tumor Cells, Cultured
10.
Cancer Lett ; 126(1): 49-57, 1998 Apr 10.
Article in English | MEDLINE | ID: mdl-9563648

ABSTRACT

In previous papers we have demonstrated that sialoadenectomy inhibited MPA-induced mammary tumorigenesis in BALB/c mice. To further explore the role of EGF in this experimental model, we evaluated its effects on mammary glands of sialoadenectomized (sialox) MPA-treated female mice and on tumor growth. MPA-treated sialox mice were injected s.c. (n = 3) or not (n = 6) with 5 microg EGF every 36 h for 45 days; MPA-treated sham-operated mice were used as controls (n = 6). Mammary glands from sialox MPA-treated mice are considerably less developed as compared with sham-operated animals. The exogenous administration of EGF restores the usual MPA-induced growth pattern of the glands, thus confirming a role for EGF either in mediating or cooperating with MPA in inducing the mammary architectural changes observed in MPA-treated mice. On the other hand, primary cultures of progestin-dependent (PD) ductal mammary adenocarcinoma in vivo tumor lines and of lobular progestin-independent (PI) tumor lines were used to evaluate the effect of EGF on tumor growth. In vitro EGF was found to stimulate cell proliferation of lobular PI tumor cells and of fibroblastic cells from both types of tumors at concentrations higher than 0.1-0.5 ng/ml and in the presence of 1-5% of charcoal-stripped fetal calf serum. Conversely, no proliferative effects were observed in ductal PD cells under the same experimental conditions, regardless of the presence of 10 nM MPA. It can be concluded that although EGF plays an important role in MPA-induced mammary carcinogenesis, it is not necessary in PD tumor growth.


Subject(s)
Adenocarcinoma/chemically induced , Carcinogens , Epidermal Growth Factor/physiology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/chemically induced , Medroxyprogesterone Acetate , Animals , Cell Division/drug effects , Female , Hyperplasia/chemically induced , Mice , Mice, Inbred BALB C , Salivary Glands/physiology , Tumor Cells, Cultured
11.
Carcinogenesis ; 19(3): 529-31, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9525291

ABSTRACT

The promoter effect of medroxyprogesterone acetate (MPA) on mammary carcinogenesis in female BALB/c mice was investigated using methylnitrosourea (MNU) as initiator. Nine out of 43 animals developed mammary carcinomas in the group treated with MNU (50 mg/kg) and MPA (administration of 40 mg every 3 months) starting 1 week after MNU administration. No tumors appeared in controls receiving only MNU or MPA during the time course of the experiment (9 months). The tumors were lobular adenocarcinomas showing different degrees of squamous differentiation with low or undetectable estrogen and progesterone receptors, and expressing epidermal growth factor receptors. These results support the hypothesis that MPA promotes the growth of MNU induced lesions.


Subject(s)
Adenocarcinoma/chemically induced , Carcinogens/toxicity , Mammary Neoplasms, Experimental/chemically induced , Medroxyprogesterone Acetate/toxicity , Methylnitrosourea/toxicity , Animals , Cocarcinogenesis , Female , Mice , Mice, Inbred BALB C
12.
J Steroid Biochem Mol Biol ; 67(4): 305-17, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9883987

ABSTRACT

The role of the insulin-like growth factors (IGFs) system was investigated in hormone-dependent (HD) and -independent (HI) in vivo lines of the medroxyprogesterone acetate (MPA)-induced mammary tumor model in Balb/c mice. IGF-II protein and message showed a three- to four-fold increase in HD lines growing in MPA-treated mice, as compared with HD tumors growing in untreated mice. Progression to a hormone-independent phenotype in all these lines was accompanied by a high constitutive expression of IGF-II. Similar IGF-I mRNA levels were detected in HD and HI lines. Both IGF-I and -II messages arose from the malignant epithelial cells, as shown by in situ hybridization studies. A significant decrease in Man-6P/type II IGF-R content was detected in HD tumors growing in MPA-treated mice as compared with HD lines growing in untreated mice. On the other hand, in HI tumors, notwithstanding high IGF-II synthesis, the levels of Man-6P/type II IGF-R remain high. Competitive inhibition and affinity labeling studies showed an almost exclusive binding of IGF-II to Man-6P/type II IGF-R on tumor membranes. The involvement of IGFs in the growth of epithelial primary cultures of the C4-HD line was evaluated. Exogenous IGF-I potentiated MPA stimulatory effect at concentrations of 50-100 ng/ml. Treatment of C4-HD cells with antisense oligodeoxynucleotides (ASODNs) to type I IGF-R and to IGF-II RNA resulted in a dose-dependent inhibition of MPA-mediated cell proliferation. The inhibition caused by IGF-II ASODNs could not be overcome by the addition of IGF-II up to 150 ng/ml. ASODNs to type I IGF-R at 40 microg/ml reduced by 75% the number of type I IGF-R; ASODNs to IGF-II at 1 microM decreased by 83% the levels of IGF-II protein. Our results provide support for the involvement of IGF-I and -II in MPA-induced mammary tumor growth by autocrine pathways.


Subject(s)
Insulin-Like Growth Factor II/physiology , Insulin-Like Growth Factor I/physiology , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/pharmacology , Receptor, IGF Type 1/physiology , Receptor, IGF Type 2/physiology , Adenocarcinoma/pathology , Animals , Base Sequence , Cell Division/drug effects , Cell Division/physiology , Female , In Situ Hybridization , Mice , Mice, Inbred BALB C , Oligonucleotides, Antisense/pharmacology , Tumor Cells, Cultured
13.
Medicina (B Aires) ; 57 Suppl 2: 55-69, 1997.
Article in Spanish | MEDLINE | ID: mdl-9567344

ABSTRACT

We have developed an experimental model in which the administration of progestins induces mammary tumors in female virgin BALB/c mice. In this paper we review the morphological and biological features of progestin-induced tumors, such as estrogen receptor (ER) and progesterone receptor (PR) patterns of expression, hormone dependence and epidermal growth factor receptors (EGF-R) we also examine our data concerning the systemic effects of medroxyprogesterone acetate (MPA) as regards its stimulating EGF synthesis in salivary glands and its subsequent increase in serum. This growth factor seems to play an important role in the induction of mammary tumors. Direct MPA proliferative effects mediated by PR were demonstrated using primary cultures of progestin-dependent (PD) mammary tumors. Antiprogestins inhibited cell growth beyond control values, suggesting that PR are involved in cell proliferation even in the absence of the ligand. Progesterone-independent (PI) tumors expressing high levels of PR and ER are also inhibited by estrogen or antiprogestin treatment, suggesting that PR are involved in the control of autonomous tumor growth. Estrogen-resistant variants may be selected which may revert to an estrogen-sensitive phenotype after several transplants in untreated mice. The similarities between the tumors obtained with this model and human breast cancer as regards morphological features, evolution and the regulation of growth control converts this model into a useful tool to explore the mechanisms related with acquisition of hormone independence and autonomous tumor growth.


Subject(s)
Adenocarcinoma/pathology , ErbB Receptors/analysis , Mammary Neoplasms, Experimental/pathology , Neoplasms, Hormone-Dependent/pathology , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Adenocarcinoma/chemically induced , Animals , Child , Disease Progression , Female , Humans , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/chemically induced , Medroxyprogesterone Acetate , Mice , Mice, Inbred BALB C
14.
Breast Cancer Res Treat ; 35(2): 173-86, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7647339

ABSTRACT

The effect of progesterone (Pg), medroxyprogesterone acetate (MPA), estradiol (E2), dihydrotestosterone (DHT) and dexamethasone (DEXA) was studied on the in vitro growth rate of a progestin-dependent (PD), estrogen-sensitive mammary tumor line originated in an MPA-treated BALB/c mouse (C4-HD), and on its estrogen-resistant variant (C4-HDR). The specificity of hormone action was further investigated using the anti-hormones RU-486 and hydroxyflutamide (FLU). Cell growth was evaluated in epithelial and fibroblast-enriched cultures using 3H-thymidine and/or autoradiography and immunocytochemistry. The results indicate that cell growth is directly stimulated by MPA and Pg at concentrations ranging from 10(-11) to 10(-7) M. RU486 prevented MPA-induced stimulation in concentrations 10 to 100 fold lower than those of MPA. When used alone, it inhibited cell proliferation only in concentrations higher than 10(-11) M. At nM concentrations, neither DEXA nor DHT stimulated 3H-thymidine uptake except DEXA at 100 nM. MPA-induced stimulation was not reverted by micromolar concentrations of FLU. As for E2 (10(-7)-10(-9) M) it prevented MPA stimulation only in cultures of estrogen-sensitive tumors. Progesterone receptors (PR) (475 +/- 115 fmoles/10(5) cells, n = 5) and estrogen receptors (ER) (ND-115 fmoles/10(5) cells, n = 5) were detected only in epithelial-enriched cultures. Serum from 7 day-MPA-treated mice induced a significant increase of 3H-thymidine uptake; an increase was also obtained with serum from untreated ovariectomized animals to which 1 nM-100 nM concentrations of MPA had been added. The stimulatory effect of the exogenous MPA was much lower than that of the serum obtained from MPA-treated animals. It is concluded that MPA stimulates cell growth of primary cultures of MPA-induced PD tumors via PR. The results provide support for a direct effect of MPA which may be mediated or potentiated by serum factors.


Subject(s)
Adenocarcinoma/pathology , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/pharmacology , Adenocarcinoma/blood , Adenocarcinoma/chemically induced , Androgen Antagonists/pharmacology , Animals , Blood Physiological Phenomena , Cell Division/drug effects , Dexamethasone/pharmacology , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Female , Flutamide/analogs & derivatives , Flutamide/pharmacology , Mammary Neoplasms, Experimental/blood , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Ovariectomy , Progesterone/pharmacology , Tumor Cells, Cultured
15.
Cancer Invest ; 13(2): 173-80, 1995.
Article in English | MEDLINE | ID: mdl-7874571

ABSTRACT

We investigated the expression of transforming growth factors beta 1 and alpha (TGF-beta 1, TGF-alpha) in hormone-responsive (MPA-R) and unresponsive (MPA-U) tumor lines obtained from medroxyprogesterone acetate (MPA)-induced mammary adenocarcinomas in BALB/c mice. The tumors were transplanted into MPA-treated and untreated mice. TGF-beta 1 gene expression was observed in the MPA-R lines growing in untreated animals, but not in MPA-treated mice. TGF-beta 1 mRNA was not detected in the MPA-U tumor lines growing in either MPA-treated or untreated animals. In MPA-R lines the levels of TGF-beta 1 expression were inversely correlated to growth rate. High-affinity TGF-beta 1 receptors were present in the MPA-R tumors. These results suggest that one of the mechanisms by which MPA exerts its proliferative effect on MPA-R tumor lines is inhibition of the expression of TGF-beta 1. Thus, the lack of expression of TGF-beta 1 in MPA-U tumors may be related to the acquisition of autonomous growth.


Subject(s)
Adenocarcinoma/metabolism , Mammary Neoplasms, Experimental/metabolism , Medroxyprogesterone Acetate/pharmacology , Neoplasms, Hormone-Dependent/metabolism , Transforming Growth Factor beta/biosynthesis , Adenocarcinoma/chemically induced , Animals , Female , Gene Expression , Mammary Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/chemically induced , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor alpha/biosynthesis , Transforming Growth Factor alpha/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
16.
Int J Cancer ; 59(2): 196-203, 1994 Oct 15.
Article in English | MEDLINE | ID: mdl-7927919

ABSTRACT

To evaluate the possible involvement of the salivary glands in the modulation of medroxyprogesterone (MPA)-induced mammary tumorigenesis, 48 sialoadenectomized virgin BALB/c female mice and 47 controls were treated with 40mg MPA depot s.c. every 3 months for 1 year. Mammary tumors developed in 11 sialoadenectomized and in 34 control mice with similar latencies. In both groups, 75% of the tumors were ductal and progestin-dependent (PD) while the remainder were lobular and progestin-independent (PI). Epidermal growth factor (EGF) levels were measured in salivary glands (SG-EGF) and serum (S-EGF) in both groups. MPA induced a significant increase in SG-EGF and in S-EGF that became evident only after 1 month of MPA treatment. No increase in S-EGF was detected in MPA-treated sialoadenectomized mice, indicating that salivary glands are the major source of S-EGF. The presence of EGF receptors (EGF-R) was investigated in ductal PD and PI tumor lines and compared with 8 PI tumor lines of lobular origin. A significant difference in EGF-R content was found between lobular and ductal tumors. No increase in EGF-R was noted when ductal tumors became autonomous. EGF-R did not correlate with tumor growth rate and there was an inverse correlation between EGF-R and steroid receptors. When the effect of sialoadenectomy on tumor growth was tested in vivo in syngeneic transplants of 2 ductal PD, 1 ductal PI and 2 lobular PI mammary adenocarcinomas, it was not found to be significant when compared with the controls. It may be concluded that SG-EGF plays an important role in the induction of mammary adenocarcinomas by MPA, while it has no significant effect on the growth of established tumors.


Subject(s)
Epidermal Growth Factor/physiology , ErbB Receptors/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/toxicity , Neoplasms, Hormone-Dependent/chemically induced , Neoplasms, Hormone-Dependent/pathology , Salivary Glands/physiology , Animals , Cell Division/physiology , Epidermal Growth Factor/blood , Epidermal Growth Factor/metabolism , ErbB Receptors/analysis , Female , Mammary Neoplasms, Experimental/ultrastructure , Mice , Mice, Inbred BALB C , Neoplasms, Hormone-Dependent/ultrastructure , Salivary Glands/metabolism , Salivary Glands/surgery
17.
Breast Cancer Res Treat ; 28(1): 29-39, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8123867

ABSTRACT

We have demonstrated that medroxyprogesterone acetate (MPA), when administered in high doses, induces mammary carcinomas in virgin female BALB/c mice. Since one of the possible explanations for this effect was its progestagenic effects, we decided to investigate whether progesterone (Pg) alone could also induce mammary adenocarcinomas in our model and if MPA at doses lower than those used to establish the model was also carcinogenic. A total of 136 mice were subdivided into 3 groups: Group 1, 44 mice were implanted s.c. with 40 mg Pg silastic pellets at the beginning of the experiment, and 6 months later with a 20 mg Pg pellet; Group 2, 45 mice were similarly treated with MPA pellets; Group 3, 47 mice were inoculated s.c. with 40 mg MPA every three months. At the end of 20 months, 9 animals had developed mammary tumors in Group 1, 18 in Group 2 and 34 in Group 3 (actuarial incidence = 28%, 58%, and 98%, respectively); tumor latency was similar in all groups: 46.2 +/- 13.1, 51.3 +/- 9.9, and 50.1 +/- 2.1 weeks, respectively. Seven (Group 1), 14 (Group 2), and 25 (Group 3) tumors were transplanted into syngeneic mice to determine progestin dependence. All tumors, except one from Group 1, were histologically characterized. In Group 1 (Pg 60 mg), 4 tumors (67%) were infiltrating lobular carcinomas and 2 were ductal carcinomas (33%). In Group 2 (MPA 60 mg), 2 tumors (14%) were lobular and 12 were ductal adenocarcinomas (86%) (Group 1 vs Group 2: p < 0.05), whereas in Group 3 (MPA 160 mg), 8 were lobular carcinomas (32%) and 17 were ductal carcinomas (68%). In syngeneic passages all lobular tumors behaved as progestin independent (PI) and ductal tumors as progestin dependent (PD). All ductal tumors, except one, expressed estrogen receptors (ER) and progesterone receptors (PR), whereas receptor expression was variable in lobular carcinomas. It can be concluded that Pg induces mostly lobular, PI mammary tumors in BALB/c female mice. The fact that most MPA-induced tumors are ductal and PD suggests that the two hormones use different carcinogenic pathways.


Subject(s)
Adenocarcinoma/chemically induced , Carcinoma, Ductal, Breast/chemically induced , Carcinoma, Lobular/chemically induced , Mammary Neoplasms, Experimental/chemically induced , Neoplasms, Hormone-Dependent/chemically induced , Progesterone/toxicity , Progestins/physiology , Adenocarcinoma/pathology , Animals , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/ultrastructure , Carcinoma, Lobular/pathology , Carcinoma, Lobular/ultrastructure , Female , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/ultrastructure , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred BALB C , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/ultrastructure , Receptors, Estrogen/physiology , Receptors, Progesterone/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...