Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biol Bull ; 244(3): 190-200, 2023 Jun.
Article in English | MEDLINE | ID: mdl-38457678

ABSTRACT

AbstractSpecies undergoing postreproductive death experience great changes in their reproductive organs, which are driven by numerous physiological processes. To assess whether apoptotic processes are involved in the dynamics of the reproductive organs of Pleurobranchaea maculata, the gonadal structure of this semelparous side-gilled sea slug was studied using light and scanning electron microscopy. Apoptotic cells at different gonadal developmental stages were detected by in situ TUNEL assay. Apoptosis was primarily focused on spermatogonia during gonadal cell proliferation, probably as a regulatory mechanism that maintains homeostasis in reproductive cells. Visible gonadal degeneration at the end of the reproductive period is accompanied by apoptosis of the basal lamina cells of the acini, suggesting that apoptotic processes are involved in the gonadal degeneration observed in P. maculata.


Subject(s)
Pleurobranchaea , Animals , Pleurobranchaea/chemistry , Apoptosis , Aplysia
2.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34977930

ABSTRACT

Luminal breast cancer (BrCa) has a favorable prognosis compared with other tumor subtypes. However, with time, tumors may evolve and lead to disease progression; thus, there is a great interest in unraveling the mechanisms that drive tumor metastasis and endocrine resistance. In this review, we focus on one of the many pathways that have been involved in tumor progression, the fibroblast growth factor/fibroblast growth factor receptor (FGFR) axis. We emphasize in data obtained from in vivo experimental models that we believe that in luminal BrCa, tumor growth relies in a crosstalk with the stromal tissue. We revisited the studies that illustrate the interaction between hormone receptors and FGFR. We also highlight the most frequent alterations found in BrCa cell lines and provide a short review on the trials that use FGFR inhibitors in combination with endocrine therapies. Analysis of these data suggests there are many players involved in this pathway that might be also targeted to decrease FGF signaling, in addition to specific FGFR inhibitors that may be exploited to increase their efficacy.


Subject(s)
Breast Neoplasms/drug therapy , Fibroblast Growth Factors/physiology , Receptors, Fibroblast Growth Factor/physiology , Receptors, Steroid/physiology , Signal Transduction/physiology , Animals , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/chemistry , Breast Neoplasms/therapy , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Estrogen Receptor alpha/analysis , Female , Fibroblast Growth Factors/genetics , Gene Amplification , Humans , Mice , Mutation , Receptor Cross-Talk/physiology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/genetics
3.
Int J Cancer ; 150(9): 1481-1496, 2022 05 01.
Article in English | MEDLINE | ID: mdl-34935137

ABSTRACT

Progesterone receptors (PRs) ligands are being tested in luminal breast cancer. There are mainly two PR isoforms, PRA and PRB, and their ratio (PRA/PRB) may be predictive of antiprogestin response. Our aim was to investigate: the impact of the PR isoform ratio on metastatic behaviour, the PR isoform ratio in paired primary tumours and lymph node metastases (LNM) and, the effect of antiprogestin/progestins on metastatic growth. Using murine and human metastatic models, we demonstrated that tumours with PRB > PRA (PRB-H) have a higher proliferation index but less metastatic ability than those with PRA > PRB (PRA-H). Antiprogestins and progestins inhibited metastatic burden in PRA-H and PRB-H models, respectively. In breast cancer samples, LNM retained the same PRA/PRB ratio as their matched primary tumours. Moreover, PRA-H LNM expressed higher total PR levels than the primary tumours. The expression of NDRG1, a metastasis suppressor protein, was higher in PRB-H compared to PRA-H tumours and was inversely regulated by antiprogestins/progestins. The binding of the corepressor SMRT at the progesterone responsive elements of the NDRG1 regulatory sequences, together with PRA, impeded its expression in PRA-H cells. Antiprogestins modulate the interplay between SMRT and AIB1 recruitment in PRA-H or PRB-H contexts regulating NDRG1 expression and thus, metastasis. In conclusion, we provide a mechanistic interpretation to explain the differential role of PR isoforms in metastatic growth and highlight the therapeutic benefit of using antiprogestins in PRA-H tumours. The therapeutic effect of progestins in PRB-H tumours is suggested.


Subject(s)
Breast Neoplasms , Cell Cycle Proteins , Intracellular Signaling Peptides and Proteins , Receptors, Progesterone , Animals , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasm Metastasis , Progesterone/pharmacology , Progestins/metabolism , Protein Isoforms/metabolism , Receptors, Progesterone/metabolism
4.
Essays Biochem ; 65(6): 951-969, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34061163

ABSTRACT

Progesterone receptors (PR) play a pivotal role in many female reproductive tissues such as the uterus, the ovary, and the mammary gland (MG). Moreover, PR play a key role in breast cancer growth and progression. This has led to the development and study of different progestins and antiprogestins, many of which are currently being tested in clinical trials for cancer treatment. Recent reviews have addressed the role of PR in MG development, carcinogenesis, and breast cancer growth. Thus, in this review, in addition to making an overview on PR action in normal and tumor breast, the focus has been put on highlighting the still unresolved topics on hormone treatment involving PR isoforms and breast cancer prognosis.


Subject(s)
Breast Neoplasms , Receptors, Progesterone , Breast/pathology , Breast Neoplasms/drug therapy , Female , Humans , Progesterone/therapeutic use , Progestins/therapeutic use , Receptors, Progesterone/therapeutic use
5.
Sci Rep ; 10(1): 15062, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934281

ABSTRACT

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Anti-Mullerian Hormone/metabolism , Estrogen Receptor alpha/biosynthesis , Neoplasm Proteins/biosynthesis , Peutz-Jeghers Syndrome/metabolism , Response Elements , Sertoli Cells/metabolism , Androgen-Insensitivity Syndrome/pathology , Animals , Cell Line , Child , Child, Preschool , Estradiol/metabolism , Female , Humans , Male , Mice , Peutz-Jeghers Syndrome/pathology , Sertoli Cells/pathology
6.
Sci Rep ; 10(1): 9507, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32528077

ABSTRACT

The superposition of male sexual characteristics in female marine gastropods (imposex) represents one of the clearest ecological examples of organotin-mediated endocrine disruption. Recent evidences suggest that signaling pathways mediated by members of the nuclear receptor superfamily, RXR and PPARγ, are involved in the development of this pseudohermaphroditic condition. Here, we identified significant differences in RXR expression in two caenogastropod species from Nuevo Gulf, Argentina, Buccinanops globulosus and Trophon geversianus, which present clear contrast in imposex incidence. In addition, B. globulosus males from a polluted and an unpolluted area showed differences in RXR expression. Conversely, PPARγ levels were similar between both analyzed species. These findings indicate specie-specific RXR and PPARγ expression, suggesting a major role of RXR in the induction of imposex.


Subject(s)
Disorders of Sex Development/metabolism , Gastropoda , Gene Expression Regulation, Developmental , Retinoid X Receptors/metabolism , Animals , Disease Susceptibility , Disorders of Sex Development/pathology , Female , Male , PPAR gamma/metabolism
7.
Int J Cancer ; 145(7): 1874-1888, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30843188

ABSTRACT

Progression to hormone-independent growth leading to endocrine therapy resistance occurs in a high proportion of patients with estrogen receptor alpha (ERα) and progesterone receptors (PR) positive breast cancer. We and others have previously shown that estrogen- and progestin-induced tumor growth requires ERα and PR interaction at their target genes. Here, we show that fibroblast growth factor 2 (FGF2)-induces cell proliferation and tumor growth through hormone-independent ERα and PR activation and their interaction at the MYC enhancer and proximal promoter. MYC inhibitors, antiestrogens or antiprogestins reverted FGF2-induced effects. LC-MS/MS identified 700 canonical proteins recruited to MYC regulatory sequences after FGF2 stimulation, 397 of which required active ERα (ERα-dependent). We identified ERα-dependent proteins regulating transcription that, after FGF2 treatment, were recruited to the enhancer as well as proteins involved in transcription initiation that were recruited to the proximal promoter. Also, among the ERα-dependent and independent proteins detected at both sites, PR isoforms A and B as well as the novel protein product PRBΔ4 were found. PRBΔ4 lacks the hormone-binding domain and was able to induce reporter gene expression from estrogen-regulated elements and to increase cell proliferation when cells were stimulated with FGF2 but not by progestins. Analysis of the Cancer Genome Atlas data set revealed that PRBΔ4 expression is associated with worse overall survival in luminal breast cancer patients. This discovery provides a new mechanism by which growth factor signaling can engage nonclassical hormone receptor isoforms such as PRBΔ4, which interacts with growth-factor activated ERα and PR to stimulate MYC gene expression and hence progression to endocrine resistance.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Fibroblast Growth Factor 2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Prognosis , Promoter Regions, Genetic , Protein Interaction Maps , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Progesterone/genetics , Survival Analysis , Xenograft Model Antitumor Assays
8.
J Pathol Clin Res ; 3(4): 227-233, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29085663

ABSTRACT

Progesterone receptors (PR) are prognostic and predictive biomarkers in hormone-dependent cancers. Two main PR isoforms have been described, PRB and PRA, that differ only in that PRB has 164 extra N-terminal amino acids. It has been reported that several antibodies empirically exclusively recognize PRA in formalin-fixed paraffin-embedded (FFPE) tissues. To confirm these findings, we used human breast cancer xenograft models, T47D-YA and -YB cells expressing PRA or PRB, respectively, MDA-MB-231 cells modified to synthesize PRB, and MDA-MB-231/iPRAB cells which can bi-inducibly express either PRA or PRB. Cells were injected into immunocompromised mice to generate tumours exclusively expressing PRA or PRB. PR isoform expression was verified using immunoblots. FFPE samples from the same tumours were studied by immunohistochemistry using H-190, clone 636, clone 16, and Ab-6 anti-PR antibodies, the latter exclusively recognizing PRB. Except for Ab-6, all antibodies displayed a similar staining pattern. Our results indicate that clones 16, 636, and the H-190 antibody recognize both PR isoforms. They point to the need for more stringency in evaluating the true specificity of purported PRA-specific antibodies as the PRA/PRB ratio may have prognostic and predictive value in breast cancer.

9.
Int J Cancer ; 136(11): 2680-92, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25363551

ABSTRACT

There is emerging interest in understanding the role of progesterone receptors (PRs) in breast cancer. The aim of this study was to investigate the proliferative effect of progestins and antiprogestins depending on the relative expression of the A (PRA) and B (PRB) isoforms of PR. In mifepristone (MFP)-resistant murine carcinomas antiprogestin responsiveness was restored by re-expressing PRA using demethylating agents and histone deacetylase inhibitors. Consistently, in two human breast cancer xenograft models, one manipulated to overexpress PRA or PRB (IBH-6 cells), and the other expressing only PRA (T47D-YA) or PRB (T47D-YB), MFP selectively inhibited the growth of PRA-overexpressing tumors and stimulated IBH-6-PRB xenograft growth. Furthermore, in cells with high or equimolar PRA/PRB ratios, which are stimulated to proliferate in vitro by progestins, and are inhibited by MFP, MPA increased the interaction between PR and the coactivator AIB1, and MFP favored the interaction between PR and the corepressor SMRT. In a PRB-dominant context in which MFP stimulates and MPA inhibits cell proliferation, the opposite interactions were observed. Chromatin immunoprecipitation assays in T47D cells in the presence of MPA or MFP confirmed the interactions between PR and the coregulators at the CCND1 and MYC promoters. SMRT downregulation by siRNA abolished the inhibitory effect of MFP on MYC expression and cell proliferation. Our results indicate that antiprogestins are therapeutic tools that selectively inhibit PRA-overexpressing tumors by increasing the SMRT/AIB1 balance at the CCND1 and MYC promoters.


Subject(s)
Cyclin D1/genetics , Mammary Neoplasms, Experimental/drug therapy , Mifepristone/pharmacology , Progestins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptors, Progesterone/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Coactivator 3/metabolism , Promoter Regions, Genetic
10.
Vitam Horm ; 93: 161-84, 2013.
Article in English | MEDLINE | ID: mdl-23810006

ABSTRACT

Hormone receptors represent the earliest biomarkers used in breast cancer not only as prognosis markers but, in addition, to decide treatment. However, mostly estrogen receptors have been used as therapeutic targets. There is compelling evidence indicating that progesterone receptors (PRs) play a hierarchical role in breast cancer growth and that they might be potentially used to improve the success of endocrine treatments. The two PR isoforms, PR-A and PR-B, play differential roles in regulating gene expression. Tumors overexpressing one or other PR isoform may respond different to endocrine treatment. In this chapter, we highlight the evidence regarding progestins as promoters or inhibitors of cell proliferation in order to understand the dual role of PR in regulating tumor growth, underscoring thus the need of biomarkers to identify which patients may benefit with an antiprogestin/progestin treatment.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Molecular Targeted Therapy , Neoplasm Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Female , Humans , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Progestins/adverse effects , Progestins/metabolism , Progestins/pharmacology , Progestins/therapeutic use , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors
11.
Medicina (B Aires) ; 72(4): 315-20, 2012.
Article in Spanish | MEDLINE | ID: mdl-22892083

ABSTRACT

In C4-HD murine mammary carcinomas and in human breast cancer T47D cells, we showed that medroxyprogesterone acetate (MPA) induces a nuclear physical association between estrogen receptor alpha (ERa) and progesterone receptors (PR). The blockade of ERa inhibits cell proliferation mediated by progestins. We hypothesized that this nuclear association between ERa/PR is necessary to trigger progestin-induced cell proliferation and tumor growth. We demonstrated that fulvestrant (FUL, ICI182.780) induced complete regression of C4-HD tumors growing with progestins. MPA treatment induced an early increase in both CCND1 and MYC expression in T47D cells. The blockade of ERa prevented the MPA-dependent transcription of both genes. Specific binding of PR/ERa was observed at the same MPA-sensitive regions at the CCND1 and MYC gene promoters after chromatin immunoprecipitation (ChIP) analysis. ICI inhibited binding of ERa to both gene regulatory sequences while PR binding was unaffected. The nuclear colocalization between both receptors in T47D cells was confirmed by: confocal microscopy, Duolink assays and co-immunoprecipitation assays. In breast cancer samples we also observed a nuclear interaction between both steroid receptors. Our results indicate that the presence of ERa interacting with activated PR at the CCND1 and MYC promoters is required to trigger progestin-induced gene transcription and cell proliferation in breast cancer cells.


Subject(s)
Carcinoma/pathology , Estradiol/analogs & derivatives , Estrogen Receptor alpha/physiology , Mammary Neoplasms, Experimental/pathology , Receptors, Progesterone/physiology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Carcinoma/chemically induced , Carcinoma/drug therapy , Cell Proliferation , Chromatin Immunoprecipitation , Cyclin D1/metabolism , Estradiol/administration & dosage , Estrogen Receptor alpha/drug effects , Female , Fulvestrant , Genes, myc , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Medroxyprogesterone Acetate/pharmacology , Murinae , Progestins/metabolism , Receptors, Progesterone/drug effects , Transcription, Genetic
12.
Cancer Res ; 72(9): 2416-27, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22396492

ABSTRACT

Synthetic progesterone used in contraception drugs (progestins) can promote breast cancer growth, but the mechanisms involved are unknown. Moreover, it remains unclear whether cytoplasmic interactions between the progesterone receptor (PR) and estrogen receptor alpha (ERα) are required for PR activation. In this study, we used a murine progestin-dependent tumor to investigate the role of ERα in progestin-induced tumor cell proliferation. We found that treatment with the progestin medroxyprogesterone acetate (MPA) induced the expression and activation of ERα, as well as rapid nuclear colocalization of activated ERα with PR. Treatment with the pure antiestrogen fulvestrant to block ERα disrupted the interaction of ERα and PR in vitro and induced the regression of MPA-dependent tumor growth in vivo. ERα blockade also prevented an MPA-induced increase in CYCLIN D1 (CCND1) and MYC expression. Chromatin immunoprecipitation studies showed that MPA triggered binding of ERα and PR to the CCND1 and MYC promoters. Interestingly, blockade or RNAi-mediated silencing of ERα inhibited ERα, but not PR binding to both regulatory sequences, indicating that an interaction between ERα and PR at these sites is necessary for MPA-induced gene expression and cell proliferation. We confirmed that nuclear colocalization of both receptors also occurred in human breast cancer samples. Together, our findings argued that ERα-PR association on target gene promoters is essential for progestin-induced cell proliferation.


Subject(s)
Breast Neoplasms/pathology , Cyclin D1/genetics , Estrogen Receptor alpha/metabolism , Genes, myc , Mammary Neoplasms, Experimental/pathology , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Growth Processes/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Fulvestrant , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Promoter Regions, Genetic , Receptors, Progesterone/genetics
13.
Cancer Res ; 71(10): 3720-31, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21464042

ABSTRACT

Fibroblast growth factor (FGF) receptor 2 (FGFR-2) polymorphisms have been associated with an increase in estrogen receptor and progesterone receptor (PR)-positive breast cancer risk; however, a clear mechanistic association between FGFR-2 and steroid hormone receptors remains elusive. In previous works, we have shown a cross talk between FGF2 and progestins in mouse mammary carcinomas. To investigate the mechanisms underlying these interactions and to validate our findings in a human setting, we have used T47D human breast cancer cells and human cancer tissue samples. We showed that medroxyprogesterone acetate (MPA) and FGF2 induced cell proliferation and activation of ERK, AKT, and STAT5 in T47D and in murine C4-HI cells. Nuclear interaction between PR, FGFR-2, and STAT5 after MPA and FGF2 treatment was also showed by confocal microscopy and immunoprecipitation. This effect was associated with increased transcription of PRE and/or GAS reporter genes, and of PR/STAT5-regulated genes and proteins. Two antiprogestins and the FGFR inhibitor PD173074, specifically blocked the effects induced by FGF2 or MPA respectively. The presence of PR/FGFR-2/STAT5 complexes bound to the PRE probe was corroborated by using NoShift transcription and chromatin immunoprecipitation of the MYC promoter. Additionally, we showed that T47D cells stably transfected with constitutively active FGFR-2 gave rise to invasive carcinomas when transplanted into NOD/SCID mice. Nuclear colocalization between PR and FGFR-2/STAT5 was also observed in human breast cancer tissues. This study represents the first demonstration of a nuclear interaction between FGFR-2 and STAT5, as PR coactivators at the DNA progesterone responsive elements, suggesting that FGFRs are valid therapeutic targets for human breast cancer treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Progesterone/metabolism , STAT5 Transcription Factor/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation
14.
Breast Cancer Res Treat ; 129(1): 49-67, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20890655

ABSTRACT

Over the past several years, we have been interested in understanding the mechanisms by which mammary carcinomas acquire hormone independence. We demonstrated that carcinoma associated fibroblasts participate in the ligand-independent activation of progesterone receptors inducing tumor growth. In this study, we used DNA microarrays to compare the gene expression profiles of tumors from the MPA mouse breast cancer model, one hormone-dependent (C4-HD) and one hormone-independent (C4-HI), using whole tumor samples or laser-captured purified stromal and epithelial cells obtained from the same tumors. The expression of selected genes was validated by immunohistochemistry and immunofluorescence assays. We identified 413 genes specifically expressed in tumor stroma. Eighty-five percent of these genes were upregulated, whereas the remaining 15% were downregulated in C4-HI relative to their expression in the C4-HD tumor stroma. Several matrix metallopeptidases were overexpressed in the C4-HI tumor microenvironment. On the other hand, 1100 genes were specifically expressed in the tumor parenchyma. Among them, the 29% were upregulated, whereas the remaining 71% were downregulated in C4-HI relative to C4-HD tumor epithelium. Steap, Pdgfc, Runx2, Cxcl9, and Sdf2 were among the genes with high expression in the C4-HI tumor parenchyma. Interestingly, Fgf2 was one of the few genes upregulated by MPA in C4-HD tumors, confirming its pivotal role in regulating tumor growth in this model. In conclusion, we demonstrate herein a gene expression profile that distinguishes both the epithelial and the stromal cells in mammary tumors with different hormone dependence, supporting the hypothesis that the tumor-associated stroma may contribute to hormone-independent tumor growth.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Carcinoma/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Mammary Neoplasms, Experimental/genetics , Medroxyprogesterone Acetate/pharmacology , Animals , Carcinoma/metabolism , Carcinoma/pathology , Cluster Analysis , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Reproducibility of Results , Stromal Cells/drug effects , Stromal Cells/metabolism
15.
Breast Cancer Res Treat ; 126(3): 621-36, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20535544

ABSTRACT

In this article, we demonstrate the expression of functional progesterone binding sites at the cell membrane in murine mammary carcinomas that are stimulated by progestins and inhibited by antiprogestins. Using confocal immunofluorescence, ligand binding and cell compartment-specific western blots, we were able to identify the presence of the classical progesterone receptors. Medroxyprogesterone acetate (MPA) and RU-486 (1 × 10(-11) and 1 × 10(-8) M) behaved as agonists activating extracellular signal-regulated kinases (ERKs) and progestin-regulated proteins, except for Cyclin D1 and Tissue factor which failed to increase with 1 × 10(-8) M RU-486, an experimental condition that allows PR to bind DNA. These results predicted a full agonist effect at low concentrations of RU-486. Accordingly, at concentrations lower than 1 × 10(-11) M, RU-486 increased cell proliferation in vitro. This effect was abolished by incubation with the ERK kinase inhibitor PD 98059 or by OH-tamoxifen. In vivo, at a daily dose of 1.2 µg/kg body weight RU-486 increased tumor growth, whereas at 12 mg/kg induces tumor regression. Our results indicate that low concentrations of MPA and RU-486 induce similar agonistic non-genomic effects, whereas RU-486 at higher concentrations may inhibit cell proliferation by genomic-induced effects. This suggests that RU-486 should be therapeutically administered at doses high enough to guarantee its genomic inhibitory effect.


Subject(s)
Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/metabolism , Mifepristone/agonists , Mifepristone/pharmacology , Progestins/agonists , Progestins/therapeutic use , Receptors, Progesterone/metabolism , Animals , Female , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Neoplasm Transplantation , Receptors, Progesterone/chemistry , Steroids/chemistry
16.
Breast Cancer Res Treat ; 126(2): 319-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20440553

ABSTRACT

Most breast carcinomas that are estrogen receptor (ER) and progesterone receptor (PR) positive respond initially to an endocrine therapy, but over time, they develop resistance (acquired hormone resistance). Others, however, fail to respond from the beginning (constitutive resistance). Overcoming hormone resistance is one of the major desirable aims in breast cancer treatment. Using the medroxyprogesterone acetate (MPA)-induced breast cancer mouse model, we have previously demonstrated that antiprogestin-responsive tumors show a higher expression level of PR isoform A (PRA) than PR isoform B (PRB), while tumors with constitutive or acquired resistance show a higher expression level of PRB. The aim of this study was to investigate whether PRA silencing in resistant tumors was due to PRA methylation. The CpG islands located in the PRA promoter and the first exon were studied by methylation-specific PCR (MSP) in six different tumors: two antiprogestin-responsive, two constitutive-resistant, and two with acquired resistance. Only in constitutive-resistant tumors, PRA expression was silenced by DNA methylation. Next, we evaluated the effect of a demethylating agent, 5-aza-2'-deoxycytidine, on PRA expression and antiprogestin responsiveness. In constitutive-resistant tumors, 5-aza-2'-deoxycytidine treatment in vitro and in vivo restored PRA expression and antiprogestin RU-486 responsiveness. Furthermore, high levels of DNA methyltransferase (Dnmts) 1 and 3b were detected in these tumors. In conclusion, our results suggest that methyltransferase inhibitors in combination with antiprogestins may be effective in the treatment of constitutive-resistant carcinomas with a high DNA methyltransferase level.


Subject(s)
DNA Methylation/drug effects , Drug Resistance, Neoplasm , Receptors, Progesterone/genetics , Animals , Apoptosis/drug effects , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Coculture Techniques , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Hormone Antagonists/pharmacology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Mitosis/drug effects , Neoplasm Transplantation , Promoter Regions, Genetic , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Transplantation, Heterologous , Tumor Burden/drug effects
17.
Medicina (B Aires) ; 70(6): 529-32, 2010.
Article in English | MEDLINE | ID: mdl-21163742

ABSTRACT

We have previously demonstrated a crosstalk between fibroblast growth factor 2 (FGF2) and progestins inducing experimental breast cancer growth. The aim of the present study was to compare the effects of FGF2 and of medroxyprogesterone acetate (MPA) on the mouse mammary glands and to investigate whether the antiprogestin RU486 was able to reverse the MPA- or FGF2-induced effects on both, mammary gland and tumor growth. We demonstrate that FGF2 administered locally induced an intraductal hyperplasia that was not reverted by RU486, suggesting that FGF2-induced effects are progesterone receptor (PR)-independent. However, MPA-induced paraductal hyperplasia was reverted by RU486 and a partial agonistic effect was observed in RU486-treated glands. Using C4-HD tumors which only grow in the presence of MPA, we showed that FGF2 administered intratumorally was able to stimulate tumor growth as MPA. The histology of FGF2-treated tumors showed different degrees of gland differentiation. RU486 inhibited both, MPA or FGF2 induced tumor growth. However, only complete regression was observed in MPA-treated tumors. Our results support the hypothesis that stromal FGF2 activates PR inducing hormone independent tumor growth.


Subject(s)
Fibroblast Growth Factor 2/antagonists & inhibitors , Hormone Antagonists/pharmacology , Mammary Glands, Animal/drug effects , Mammary Neoplasms, Animal/drug therapy , Medroxyprogesterone Acetate/antagonists & inhibitors , Mifepristone/pharmacology , Animals , Disease Models, Animal , Female , Hyperplasia/chemically induced , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/chemically induced , Mice
18.
Breast Cancer Res Treat ; 123(3): 709-24, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20012353

ABSTRACT

To evaluate the extent to which each estrogen receptor (ER) subtype contributes to the stimulation or to the inhibition of mammary tumor growth, we evaluated the effects of specific agonists in MC4-L2 cells, which are stimulated by 17ß-estradiol (E(2)), and in mammary carcinomas of the MPA mouse breast cancer model, which are inhibited by E(2). Both express ERα and ERß. In MC4-L2 cells, 4,4',4"-(4-propyl-(1H)-pyrazole-1,3,5-triyl)trisphenol (PPT; ERα agonist) and (4-hydroxy-phenyl)-propionitrile (DPN; ERß agonist) stimulated cell proliferation, whereas the opposite occurred in C4-HI primary cultures. The inhibitory effect was associated with a decrease in ERα and cyclin D1 expression and an increase in progesterone receptor (PR) expression as well as in the Bax/Bcl-xl ratio. In vivo, mice carrying C4-HI or 32-2-HI tumors were treated with E(2), PPT or DPN (3 mg/kg/day) or with vehicle. PPT and DPN inhibited tumor size, as did E(2), during the first 72 h. After a few days, DPN-treated tumors started to grow again, while PPT-treated tumors remained quiescent for a longer period of time. A pronounced decrease in the mitotic index and an increase in the apoptotic index was associated with tumor regression. All treated tumors showed: (a) an increase in integrin α6 and Bax expression, (b) an increased stromal laminin redistribution, and (c) a decrease in ERα, Bcl-xl and Bcl-2 expression (P < 0.001). Apoptosis-inducing factor (Aif) expression was increased in DPN-treated tumors, while active caspase 9 was up-regulated in PPT-treated mice, demonstrating the involvement of the intrinsic apoptotic pathway in estrogen-induced regression in this model. In conclusion, our data indicate that although there may be some preferences for activation pathways by the different agonists, the stimulatory or inhibitory effects triggered by estrogens are cell-context dependent rather than ER isoform dependent.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Nitriles/pharmacology , Phenols/pharmacology , Pyrazoles/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 9 , Cell Proliferation/drug effects , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Mice , Mice, Inbred BALB C , Receptors, Progesterone/metabolism , Time Factors , Tumor Burden , Tumor Cells, Cultured , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
19.
J Cell Physiol ; 219(2): 477-84, 2009 May.
Article in English | MEDLINE | ID: mdl-19194992

ABSTRACT

Breast cancer is the most frequent cancer in women. However, in vivo hormone receptor positive and metastatic models are scarce. The aim of the present manuscript was to assess if the novel steroid receptor positive human cell lines IBH-4, IBH-6, and IBH-7 developed in our laboratory from primary infiltrant ductal carcinomas are good models to study in vivo human breast cancer. Cell lines or tumors were inoculated to nude mice in the presence or absence of hormone supplementation. Growth was analyzed by ANOVA followed by Tukey-Kramer's test. Steroid hormone expression was assessed by immunohistochemistry and Western blotting. The histology of the tumors was analyzed. IBH-4 and IBH-6 cells were inoculated to nude mice and 100% of the injected mice developed tumors in the presence or absence of hormone treatment, although tamoxifen inhibited growth. IBH-4 and IBH-6 cell lines in vivo gave rise to poorly differentiated carcinomas with areas of solid growth and sarcomatoid areas showing no morphological signs of epithelial differentiation. Distinct features of malignancy were observed. IBH-7 tumors in animals receiving estradiol were semi-differentiated adenocarcinomas. IBH-7 cells grew only in the presence of estradiol, but even with hormone addition, the tumor take was 20%. These tumors metastasized to the uterus and lung and vascular tumor emboli were evident. IBH-7 tumors were invasive and able to break through the peritoneum. As a conclusion, IBH-4 and IBH-6 are good models for studying tumor progression, whereas IBH-7 is a good model for tumor take, being metastatic and strictly estrogen-dependent.


Subject(s)
Breast Neoplasms , Cell Line, Tumor/physiology , Mice, Nude , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor/drug effects , Estrogen Antagonists/pharmacology , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Transplantation , Tamoxifen/pharmacology , Transplantation, Heterologous
20.
Endocr Relat Cancer ; 16(2): 333-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19190078

ABSTRACT

More than 60% of all breast neoplasias are ductal carcinomas expressing estrogen (ER) and progesterone receptors (PR). By contrast, most of the spontaneous, chemically or mouse mammary tumor virus induced tumors, as well as tumors arising in genetically modified mice do not express hormone receptors. We developed a model of breast cancer in which the administration of medroxyprogesterone acetate to BALB/c female mice induces mammary ductal carcinomas with a mean latency of 52 weeks and an incidence of about 80%. These tumors are hormone-dependent (HD), metastatic, express both ER and PR, and are maintained by syngeneic transplants. The model has been further refined to include mammary carcinomas that evolve through different stages of hormone dependence, as well as several hormone-responsive cell lines. In this review, we describe the main features of this tumor model, highlighting the role of PR as a trigger of key signaling pathways mediating tumor growth. In addition, we discuss the relevance of this model in comparison with other presently used breast cancer models pointing out its advantages and limitations and how, this model may be suitable to unravel key questions in breast cancer.


Subject(s)
Carcinoma, Ductal/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasms, Hormone-Dependent/metabolism , Receptors, Progesterone/physiology , Animals , Antineoplastic Agents, Hormonal/toxicity , Female , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred BALB C , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...