Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 337
Filter
1.
Nucleic Acids Res ; 49(22): 12716-12731, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34850111

ABSTRACT

Here, we report that in T47D breast cancer cells 50 pM progestin is sufficient to activate cell cycle entry and the progesterone gene expression program. At this concentration, equivalent to the progesterone blood levels found around the menopause, progesterone receptor (PR) binds only to 2800 genomic sites, which are accessible to ATAC cleavage prior to hormone exposure. These highly accessible sites (HAs) are surrounded by well-organized nucleosomes and exhibit breast enhancer features, including estrogen receptor alpha (ERα), higher FOXA1 and BRD4 (bromodomain containing 4) occupancy. Although HAs are enriched in RAD21 and CTCF, PR binding is the driving force for the most robust interactions with hormone-regulated genes. HAs show higher frequency of 3D contacts among themselves than with other PR binding sites, indicating colocalization in similar compartments. Gene regulation via HAs is independent of classical coregulators and ATP-activated remodelers, relying mainly on MAP kinase activation that enables PR nuclear engagement. HAs are also preferentially occupied by PR and ERα in breast cancer xenografts derived from MCF-7 cells as well as from patients, indicating their potential usefulness as targets for therapeutic intervention.


Subject(s)
Breast Neoplasms/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Progestins/physiology , Animals , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Chromatin , Estrogen Receptor alpha/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System , MCF-7 Cells , Mice , Promegestone/pharmacology , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
2.
Eur J Contracept Reprod Health Care ; 26(3): 184-194, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33645377

ABSTRACT

PURPOSE: To evaluate the pharmacokinetics and pharmacodynamics of oestriol (E3) and trimegestone (TMG) in healthy women after application of three different vaginal rings over 21 days. The vaginal rings had a nominal delivery rate of 0.413/0.050 mg/day (Test 1), 0.311/0.090 mg/day (Test 2) and 0.209/0.137 mg/day (Test 3) E3/TMG. METHODS: Thirty-five healthy women were randomised to receive a single application of Test 1, 2 or 3 (Clinical Trial NCT03343912). The E3 and TMG plasma concentration was determined by LC-MS/MS. Oestradiol (E2) and progesterone (PG) serum concentrations, and bleeding patern were determined as pharmacodynamic parameters. Safety was assessed by evaluation of adverse events and local tolerability. RESULTS: The total and maximum exposure of E3 and TMG increased in a proportional ratio to dose. However, not in a magnitude which was expected from the dose differences for E3. During Test 2 and 3 treatment all E2 and PG values remained on a well suppressed level until end of treatment. E2 and PG serum levels increased distinctly earlier after ring removal with Test 1 compared to Test 2 and 3. Test 3 achieved 95.24% of "no bleeding" days under treatment followed by Test 1 (91.67%), and Test 2 (86.15%). CONCLUSIONS: The Test 3 formulation presented the best dose combination of E3/TMG for contraception. Moreover, all vaginal rings were well tolerated.


Subject(s)
Contraceptive Agents, Female/administration & dosage , Contraceptive Devices, Female , Estriol/pharmacology , Estriol/pharmacokinetics , Estrogens/metabolism , Promegestone/analogs & derivatives , Administration, Intravaginal , Adult , Chromatography, Liquid , Estradiol/blood , Estrogens/blood , Female , Humans , Progesterone/blood , Promegestone/pharmacokinetics , Promegestone/pharmacology , Tandem Mass Spectrometry
3.
Nucleic Acids Res ; 47(20): 10645-10661, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31598691

ABSTRACT

The glucocorticoid and progesterone receptors (GR and PR) are closely related members of the steroid receptor family. Despite sharing similar structural and functional characteristics; the cognate hormones display very distinct physiological responses. In mammary epithelial cells, PR activation is associated with the incidence and progression of breast cancer, whereas the GR is related to growth suppression and differentiation. Despite their pharmacological relevance, only a few studies have compared GR and PR activities in the same system. Using a PR+/GR+ breast cancer cell line, here we report that either glucocorticoid-free or dexamethasone (DEX)-activated GR inhibits progestin-dependent gene expression associated to epithelial-mesenchymal-transition and cell proliferation. When both receptors are activated with their cognate hormones, PR and GR can form part of the same complex according to co-immunoprecipitation, quantitative microscopy and sequential ChIP experiments. Moreover, genome-wide studies in cells treated with either DEX or R5020, revealed the presence of several regions co-bound by both receptors. Surprisingly, GR also binds novel genomic sites in cells treated with R5020 alone. This progestin-induced GR binding was enriched in REL DNA motifs and located close to genes coding for chromatin remodelers. Understanding GR behavior in the context of progestin-dependent breast cancer could provide new targets for tumor therapy.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genome, Human , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Base Sequence , Binding Sites , Breast Neoplasms/pathology , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucocorticoids/pharmacology , Humans , Progestins/pharmacology , Promegestone/pharmacology , Protein Binding/drug effects , Transcription, Genetic/drug effects
4.
Exp Cell Res ; 382(1): 111433, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31100306

ABSTRACT

Autophagy is an evolutionary conserved, self-eating process that targets cellular constituents for lysosomal degradation. Transcription factor EB (TFEB) is a master regulator of autophagy by inducing the expression of genes involved in autophagic and lysosomal degradation. In breast cancer, ligand-activated progesterone receptor has been reported to influence cancer development by manipulating the autophagy pathway. However, understanding of the mechanism that underlies this autophagic response remains limited. Herein, we report that prolonged treatment with progestin R5020 upregulates autophagy in MCF-7 human breast cancer cells via a novel interplay between progesterone receptor B (PRB) and TFEB. R5020 upregulates TFEB gene expression and protein levels in a PRB-dependent manner. Additionally, R5020 enhances the co-recruitment of PRB and TFEB to each other to facilitate TFEB nuclear localization. Once in the nucleus, TFEB induces the expression of autophagy and lysosomal genes to potentiate autophagy. Together, our findings highlight a novel functional connection between ligand-activated PRB and TFEB to modulate autophagy in MCF-7 breast cancer cells. As breast cancer development is controlled by autophagy, the progestin-PRB-TFEB transduction pathway warrants future attention as a potential therapeutic target in cancer therapy.


Subject(s)
Adenocarcinoma/genetics , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Breast Neoplasms/genetics , Neoplasm Proteins/physiology , Receptors, Progesterone/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Autophagosomes/metabolism , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysosomes/metabolism , MCF-7 Cells , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Promegestone/pharmacology , Protein Interaction Mapping , Protein Transport/drug effects , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Transcriptional Activation
5.
Oncogene ; 38(15): 2722-2735, 2019 04.
Article in English | MEDLINE | ID: mdl-30538295

ABSTRACT

Progesterone and its receptor, PR, are essential for uterine leiomyoma (LM, a.k.a., fibroid) tumorigenesis, but the underlying cellular and molecular mechanisms remain unclear. The receptor activator of NF-κB (RANKL) was recently identified as a novel progesterone/PR-responsive gene that plays an important role in promoting LM growth. Here, we used RANKL as a representative gene to investigate how steroid hormone, genetic, and epigenetic signals are integrated to regulate LM stem cell (LSC) function. We demonstrated that RANKL specifically upregulates LSC proliferation through activation of Cyclin D1. RANKL gene transcription was robustly induced by the progesterone agonist R5020, leading to a dramatically higher RANKL expression in LM compared to adjacent myometrial (MM) tissue. MethylCap-Seq revealed a differentially methylated region (DMR) adjacent to the distal PR-binding site (PRBS) 87 kb upstream of the RANKL transcription start site. Hypermethylation of the DMR inhibited recruitment of PR to the adjacent PRBS. Luciferase assays indicated that the DMR and distal PRBS constitute a novel RANKL distal regulatory element that actively regulates RANKL expression. Furthermore, MED12 physically interacts with PR in LM tissue. The interaction between MED12 and PR, binding of PR and MED12 to PRBS, and RANKL gene expression are significantly higher in LM containing a distinct MED12 mutation (G44D) than in LM with wild-type MED12. In summary, our findings suggest that DNA methylation and MED12 mutation together constitute a complex regulatory network that affects progesterone/PR-mediated RANKL gene expression, with an important role in activating stem cell proliferation and fibroid tumor development.


Subject(s)
Cell Proliferation/genetics , DNA Methylation/genetics , Leiomyoma/genetics , Mediator Complex/genetics , RANK Ligand/genetics , Receptors, Progesterone/genetics , Stem Cells/pathology , Uterine Neoplasms/genetics , Adult , Cell Proliferation/drug effects , DNA Methylation/drug effects , Female , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Leiomyoma/drug therapy , Middle Aged , Progesterone/genetics , Promegestone/pharmacology , Transcription Initiation Site/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Uterine Neoplasms/drug therapy
6.
Endocrinology ; 159(10): 3581-3595, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30203004

ABSTRACT

Progesterone is a steroid hormone that plays an important role in the breast. Progesterone exerts its action through binding to progesterone receptor (PR), a transcription factor. Deregulation of the progesterone signaling pathway is implicated in the formation, development, and progression of breast cancer. Next-generation selective progesterone receptor modulators (SPRMs) have potent antiprogestin activity and are selective for PR, reducing the off-target effects on other nuclear receptors. To date, there is limited information on how the newer generation of SPRMs, specifically telapristone acetate (TPA), affect PR function at the molecular level. In this study, T47D breast cancer cells were used to investigate the molecular mechanism by which TPA antagonizes PR action. Global profiling of the PR cistrome and interactome was done with chromatin immunoprecipitation sequencing (ChIP-seq) and rapid immunoprecipitation mass spectrometry. Validation studies were done on key genes and interactions. Our results demonstrate that treatment with the progestin (R5020) alone resulted in robust PR recruitment to the chromatin, and addition of TPA reduced PR recruitment globally. TPA significantly changed coregulator recruitment to PR compared with R5020. Upon conservative analysis, three proteins (TRPS1, LASP1, and AP1G1) were identified in the R5020+TPA-treated group. Silencing TRPS1 with small interfering RNA increased PR occupancy to the known PR regulatory regions and attenuated the inhibition of gene expression after TPA treatment. TRPS1 silencing alleviated the inhibition of proliferation by TPA. In conclusion, TPA decreases PR occupancy on chromatin and recruits coregulators such as TRPS1 to the PR complex, thereby regulating PR target gene expression and associated cellular responses.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Norpregnadienes/pharmacology , Receptors, Progesterone/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , Promegestone/pharmacology , Protein Binding , Receptors, Progesterone/metabolism , Repressor Proteins , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Biomaterials ; 179: 96-108, 2018 10.
Article in English | MEDLINE | ID: mdl-29980078

ABSTRACT

Hormones play an important role in normal and diseased breast tissue development. However, they can also disrupt cell-matrix interactions and their role in extracellular matrix reorganization during epithelial morphogenesis remains poorly understood, partly due to a lack of sensitive approaches for matrix characterization. Here, we assess the hormonal regulation of matrix reorganization in a three-dimensional (3D) breast tissue culture model using a novel metric, i.e., 3D directional variance, to characterize the 3D organization of collagen fibers visualized via high-resolution, second harmonic generation imaging. This metric enables resolving and quantifying patterns of spatial organization throughout the matrix surrounding epithelial structures treated with 17ß-estradiol (E2) alone, and E2 in combination with either promegestone, a progestogen, or prolactin. Addition of promegestone results in the most disorganized fibers, while the E2 alone treatment leads to the most organized ones. Location-dependent organization mapping indicates that only the prolactin treatment leads to significant heterogeneities in the regional organization of collagen fibers, with higher levels of alignment observed at the end of the elongated epithelial structures. The observed collagen organization patterns for all groups persist for tens of micrometers. In addition, a comparison between 3D directional variance and typical 2D analysis approaches reveals an improved sensitivity of the 3D metric to identify organizational heterogeneities and differences among treatment groups. These results demonstrate that 3D directional variance is sensitive to subtle changes in the extracellular micro-environment and has the potential to elucidate reciprocal cell-matrix interactions in the context of numerous applications involving the study of normal and diseased tissue morphogenesis.


Subject(s)
Breast/drug effects , Breast/metabolism , Collagen/chemistry , Estradiol/pharmacology , Female , Humans , Progestins/pharmacology , Prolactin/pharmacology , Promegestone/pharmacology
8.
Reprod Sci ; 25(4): 598-608, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28820025

ABSTRACT

Progesterone helps maintain cervical structure during pregnancy via the progesterone receptor (PR). Two PR isoforms exist, PR-A and PR-B, which have overlapping as well as isoform-specific target genes. During late gestation, leukocytes infiltrate the cervical stroma accompanied by increased cervical cytokine levels, resembling an inflammatory process. We examined interleukin (IL)-1ß regulation of the expression of PR-A, PR-B, and genes governing prostaglandin synthesis in human cervical fibroblasts (HCFs). Since progesterone has been shown to exert anti-inflammatory actions, we also examined the capacity of progesterone (R5020) to ameliorate the actions of IL-1ß in HCFs. Interleukin-1ß induced both PR-A and PR-B mRNA in HCFs. Interleukin-1ß induced a rapid and transient loss of both PR-A and PR-B protein, followed by a latent (24 hours) increase in both PR isoforms. R5020 negated the IL-1ß-induced increase in PR-A and PR-B mRNA and protein as well as the rapid IL-1ß-induced downregulation of nuclear PR. Interleukin-1ß induced prostaglandin G/H synthase-2 (PGHS-2), but not prostaglandin G/H synthase-1 (PGHS-1), as well as prostaglandin E synthase-1 (PGES-1), but not prostaglandin F synthase (PGFS). R5020 did not ameliorate IL-1ß induction of PGHS-2 or PGES-1. Blockade of prostaglandin synthesis (indomethacin) prevented both the IL-1ß-induced increase in PR mRNA and the acute decrease in PR-A and PR-B protein, implicating a role for prostaglandins in regulating PR expression in HCFs. Although progesterone may function to maintain PR expression in a milieu of increasing cytokines in the late gestation human cervix, it does not exert an anti-inflammatory role with regard to prostaglandin E2 (PGE2) production.


Subject(s)
Cervix Uteri/metabolism , Fibroblasts/metabolism , Inflammation/metabolism , Interleukin-1beta/pharmacology , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Adult , Cervix Uteri/drug effects , Dinoprostone/metabolism , Female , Fibroblasts/drug effects , Humans , Middle Aged , Promegestone/pharmacology
9.
BMC Cancer ; 16: 326, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27215412

ABSTRACT

BACKGROUND: The synthesis of specific, potent progesterone antagonists adds potential agents to the breast cancer prevention and treatment armamentarium. The identification of individuals who will benefit from these agents will be a critical factor for their clinical success. METHODS: We utilized telapristone acetate (TPA; CDB-4124) to understand the effects of progesterone receptor (PR) blockade on proliferation, apoptosis, promoter binding, cell cycle progression, and gene expression. We then identified a set of genes that overlap with human breast luteal-phase expressed genes and signify progesterone activity in both normal breast cells and breast cancer cell lines. RESULTS: TPA administration to T47D cells results in a 30 % decrease in cell number at 24 h, which is maintained over 72 h only in the presence of estradiol. Blockade of progesterone signaling by TPA for 24 h results in fewer cells in G2/M, attributable to decreased expression of genes that facilitate the G2/M transition. Gene expression data suggest that TPA affects several mechanisms that progesterone utilizes to control gene expression, including specific post-translational modifications, and nucleosomal organization and higher order chromatin structure, which regulate access of PR to its DNA binding sites. CONCLUSIONS: By comparing genes induced by the progestin R5020 in T47D cells with those increased in the luteal-phase normal breast, we have identified a set of genes that predict functional progesterone signaling in tissue. These data will facilitate an understanding of the ways in which drugs such as TPA may be utilized for the prevention, and possibly the therapy, of human breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Gene Regulatory Networks/drug effects , Norpregnadienes/pharmacology , Promegestone/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Oligonucleotide Array Sequence Analysis/methods
10.
Fertil Steril ; 105(4): 1102-10, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26776909

ABSTRACT

OBJECTIVE: To characterize the efficacy of mifepristone treatment on extracellular matrix (ECM) production in leiomyomas. DESIGN: Laboratory study. SETTING: University research laboratory. PATIENT(S): None. INTERVENTION(S): Treatment of human immortalized two-dimensional (2D) and three-dimensional (3D) leiomyoma and myometrial cells with mifepristone and the progestin promegestone (R5020). MAIN OUTCOME MEASURE(S): Expression of COL1A1, fibronectin, versican variant V0, and dermatopontin in treated leiomyoma cells by Western blot analysis and confirmatory immunohistochemistry staining of treated 3D cultures. RESULT(S): Treatment with progestin stimulated production of COL1A1, fibronectin, versican, and dermatopontin. Mifepristone treatment inhibited protein production of these genes, most notably with versican expression. Combination treatment with both the agonist and antagonist further inhibited protein expression of these genes. Immunohistochemistry performed on 3D cultures demonstrated generalized inhibition of ECM protein concentration. CONCLUSION(S): Our study demonstrated that the progesterone agonist R5020 directly stimulated extracellular matrix components COL1A1, fibronectin, versican, and dermatopontin production in human leiomyoma cells. Progesterone antagonist mifepristone decreased protein production of these genes to levels comparable with untreated leiomyoma cells.


Subject(s)
Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Hormone Antagonists/pharmacology , Leiomyoma/metabolism , Mifepristone/pharmacology , Uterine Neoplasms/metabolism , Cell Line, Transformed , Dose-Response Relationship, Drug , Extracellular Matrix/genetics , Female , Humans , Leiomyoma/genetics , Leiomyoma/pathology , Promegestone/pharmacology , Tumor Cells, Cultured , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
11.
Mol Endocrinol ; 30(2): 158-72, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26652902

ABSTRACT

Progesterone receptor (PR) function is altered by cell signaling, but the mechanisms of kinase-specific regulation are not well defined. To examine the role of cell signaling in the regulation of PR transcriptional activity, we have utilized a previously developed mammalian-based estrogen-response element promoter array cell model and automated cell imaging and analysis platform to visualize and quantify effects of specific kinases on different mechanistic steps of PR-mediated target gene activation. For these studies, we generated stable estrogen-response element array cell lines expressing inducible chimeric PR that contains a swap of the estrogen receptor-α DNA-binding domain for the DNA-binding domain of PR. We have focused on 2 kinases important for steroid receptor activity: cyclin-dependent kinase 2 and DNA-dependent protein kinase. Treatment with either a Cdk1/2 inhibitor (NU6102) or a DNA-dependent protein kinase inhibitor (NU7441) decreased hormone-mediated chromatin decondensation and transcriptional activity. Further, we observed a quantitative reduction in the hormone-mediated recruitment of select coregulator proteins with NU6102 that is not observed with NU7441. In parallel, we determined the effect of kinase inhibition on hormone-mediated induction of primary and mature transcripts of endogenous genes in T47D breast cancer cells. Treatment with NU6102 was much more effective than NU7441, in inhibiting induction of PR target genes that exhibit a rapid increase in primary transcript expression in response to hormone. Taken together, these results indicate that the 2 kinases regulate PR transcriptional activity by distinct mechanisms.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , DNA-Activated Protein Kinase/metabolism , Receptors, Progesterone/metabolism , Transcription, Genetic , Chromones/pharmacology , Gonanes/pharmacology , HeLa Cells , Humans , Mediator Complex/metabolism , Mifepristone/pharmacology , Models, Biological , Morpholines/pharmacology , Promegestone/pharmacology , Purines/pharmacology , Recombinant Proteins/metabolism , Transcription, Genetic/drug effects
12.
Reprod Sci ; 23(3): 302-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26423601

ABSTRACT

Uterine leiomyoma are a common benign pelvic tumors composed of modified smooth muscle cells and a large amount of extracellular matrix (ECM). The proteoglycan composition of the leiomyoma ECM is thought to affect pathophysiology of the disease. To test this hypothesis, we examined the abundance (by immunoblotting) and expression (by quantitative real-time polymerase chain reaction) of the proteoglycans biglycan, decorin, and versican in leiomyoma and normal myometrium and determined whether expression is affected by steroid hormones and menstrual phase. Leiomyoma and normal myometrium were collected from women (n = 17) undergoing hysterectomy or myomectomy. In vitro studies were performed on immortalized leiomyoma (UtLM) and normal myometrial (hTERT-HM) cells with and without exposure to estradiol and progesterone. In leiomyoma tissue, abundance of decorin messenger RNA (mRNA) and protein were 2.6-fold and 1.4-fold lower, respectively, compared with normal myometrium. Abundance of versican mRNA was not different between matched samples, whereas versican protein was increased 1.8-fold in leiomyoma compared with myometrium. Decorin mRNA was 2.4-fold lower in secretory phase leiomyoma compared with proliferative phase tissue. In UtLM cells, progesterone decreased the abundance of decorin mRNA by 1.3-fold. Lower decorin expression in leiomyoma compared with myometrium may contribute to disease growth and progression. As decorin inhibits the activity of specific growth factors, its reduced level in the leiomyoma cell microenvironment may promote cell proliferation and ECM deposition. Our data suggest that decorin expression in leiomyoma is inhibited by progesterone, which may be a mechanism by which the ovarian steroids affect leiomyoma growth and disease progression.


Subject(s)
Decorin/biosynthesis , Leiomyoma/metabolism , Myometrium/metabolism , Proteoglycans/biosynthesis , Uterine Neoplasms/metabolism , Adult , Cell Line, Transformed , Cell Line, Tumor , Decorin/antagonists & inhibitors , Estradiol/pharmacology , Female , Humans , Leiomyoma/physiopathology , Middle Aged , Myometrium/drug effects , Myometrium/physiopathology , Progesterone/pharmacology , Promegestone/pharmacology , Proteoglycans/antagonists & inhibitors , Uterine Neoplasms/physiopathology
13.
Oncotarget ; 6(32): 33146-64, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26356672

ABSTRACT

Overexpression of the progesterone receptor (PR) isoform A (PR-A) is a negative prognosticator for estrogen receptor (ER)-positive breast cancer but in vitro studies have implicated PR-B in progestin-induced invasiveness. As estrogen is known to suppress invasiveness and tumor progression and as the in vitro studies were conducted in models that either lacked ER or excluded estrogen, we examined the role of PR isoforms in the context of estrogen signaling. Estrogen (< 0.01nM) strongly suppressed invasiveness in various ER+ model cell lines. At low (< 1nM) concentrations, progestins completely abrogated inhibition of invasiveness by estrogen. It was only in a higher (5 nM - 50 nM) concentration range that progestins induced invasiveness in the absence of estrogen. The ability of low dose progestins to rescue invasiveness from estrogen regulation was exclusively mediated by PR-A, whereas PR-B mediated the estrogen-independent component of progestin-induced invasiveness. Overexpression of PR-A lowered the progestin concentration needed to completely rescue invasiveness. Among estrogen-regulated genes, progestin/PR-A counter-regulated a distinctive subset, including breast tumor progression genes (e.g., HES1, PRKCH, ELF5, TM4SF1), leading to invasiveness. In this manner, at relatively low hormone concentrations (corresponding to follicular stage and post-menopausal breast tissue or plasma levels), progesterone influences breast cancer cell invasiveness by rescuing it from estrogen regulation via PR-A, whereas at higher concentrations the hormone also induces invasiveness independent of estrogen signaling, through PR-B. The findings point to a direct functional link between PR-A and progression of luminal breast cancer in the context of the entire range of pre- and post-menopausal plasma and breast tissue hormone levels.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Estradiol/pharmacology , Female , HEK293 Cells , Humans , Neoplasm Invasiveness , Postmenopause/metabolism , Premenopause/metabolism , Promegestone/pharmacology , Protein Isoforms , Signal Transduction
14.
Clin Cancer Res ; 21(5): 1063-70, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25520392

ABSTRACT

PURPOSE: To investigate whether longitudinal functional PET imaging of mammary tumors using the radiopharmaceuticals [(18)F]FDG (to measure glucose uptake), [(18)F]FES [to measure estrogen receptor (ER) levels], or [(18)F]FFNP [to measure progesterone receptor (PgR) levels] is predictive of response to estrogen-deprivation therapy. EXPERIMENTAL DESIGN: [(18)F]FDG, [(18)F]FES, and [(18)F]FFNP uptake in endocrine-sensitive and -resistant mammary tumors was quantified serially by PET before ovariectomy or estrogen withdrawal in mice, and on days 3 and 4 after estrogen-deprivation therapy. Specificity of [(18)F]FFNP uptake in ERα(+) mammary tumors was determined by competition assay using unlabeled ligands for PgR or glucocorticoid receptor (GR). PgR expression was also assayed by immunohistochemistry (IHC). RESULTS: The levels of [(18)F]FES and [(18)F]FDG tumor uptake remained unchanged in endocrine-sensitive tumors after estrogen-deprivation therapy compared with those at pretreatment. In contrast, estrogen-deprivation therapy led to a reduction in PgR expression and [(18)F]FFNP uptake in endocrine-sensitive tumors, but not in endocrine-resistant tumors, as early as 3 days after treatment; the changes in PgR levels were confirmed by IHC. Unlabeled PgR ligand R5020 but not GR ligand dexamethasone blocked [(18)F]FFNP tumor uptake, indicating that [(18)F]FFNP bound specifically to PgR. Therefore, a reduction in FFNP tumor to muscle ratio in mammary tumors predicts sensitivity to estrogen-deprivation therapy. CONCLUSIONS: Monitoring the acute changes in ERα activity by measuring [(18)F]FFNP uptake in mammary tumors predicts tumor response to estrogen-deprivation therapy. Longitudinal noninvasive PET imaging using [(18)F]FFNP is a robust and effective approach to predict tumor responsiveness to endocrine treatment.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Receptors, Progesterone/metabolism , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/pharmacology , Biomarkers , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Diagnostic Imaging , Disease Models, Animal , Drug Resistance, Neoplasm , Estrogen Receptor alpha , Female , Fluorodeoxyglucose F18 , Humans , Ligands , Mammary Neoplasms, Experimental , Mice , Positron-Emission Tomography , Promegestone/pharmacology , Tomography, X-Ray Computed
15.
J Cell Physiol ; 230(7): 1594-606, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25536295

ABSTRACT

We previously encountered regulatory processes wherein dihydrotestosterone (DHT) exerted its inhibitory effect on parathyroid hormone-related protein (PTHrP) gene repression through the estrogen receptor (ER)α, but not the androgen receptor (AR), in breast cancer MCF-7 cells. Here, we investigated whether such aberrant ligand-nuclear receptor (NR) interaction is present in prostate cancer LNCaP cells. First, we confirmed that LNCaP cells expressed large amounts of AR at negligible levels of ERα/ß or progesterone receptor. Both suppression of PTHrP and activation of prostate-specific antigen genes were observed after independent administration of 17ß-estradiol (E2), DHT, or R5020. Consistent with the notion that the LNCaP AR lost its ligand specificity due to a mutation (Thr-Ala877), experiments with siRNA targeting the respective NR revealed that the AR monopolized the role of the mediator of shared hormone-dependent regulation, which was invariably associated with nuclear translocation of this mutant AR. Microarray analysis of gene regulation by DHT, E2, or R5020 disclosed that more than half of the genes downstream of the AR (Thr-Ala877) overlapped in the LNCaP cells. Of particular interest, we realized that the AR (wild-type [wt]) and AR (Thr-Ala877) were equally responsible for the E2-AR interactions. Fluorescence microscopy experiments demonstrated that both EGFP-AR (wt) and EGFP-AR (Thr-Ala877) were exclusively localized within the nucleus after E2 or DHT treatment. Furthermore, reporter assays revealed that some other cancer cells exhibited aberrant E2-AR (wt) signaling similar to that in the LNCaP cells. We herein postulate the presence of entangled interactions between wt AR and E2 in certain hormone-sensitive cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Estradiol/metabolism , Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Active Transport, Cell Nucleus/physiology , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mutation , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Promegestone/pharmacology , Receptors, Androgen/genetics
16.
PLoS One ; 9(6): e98404, 2014.
Article in English | MEDLINE | ID: mdl-24897521

ABSTRACT

Despite the pleiotropic effects of the progesterone receptor in breast cancer, the molecular mechanisms in play remain largely unknown. To gain a global view of the PR-orchestrated networks, we used next-generation sequencing to determine the progestin-regulated transcriptome in T47D breast cancer cells. We identify a large number of PR target genes involved in critical cellular programs, such as regulation of transcription, apoptosis, cell motion and angiogenesis. Integration of the transcriptomic data with the PR-binding profiling of hormonally treated cells identifies numerous components of the small-GTPases signaling pathways as direct PR targets. Progestin-induced deregulation of the small GTPases may contribute to the PR's role in mammary tumorigenesis. Transcript expression analysis reveals significant expression changes of specific transcript variants in response to the extracellular hormonal stimulus. Using the NET1 gene as an example, we show that the PR can dictate alternative promoter usage leading to the upregulation of an isoform that may play a role in metastatic breast cancer. Future studies should aim to characterize these selectively regulated variants and evaluate their clinical utility in prognosis and targeted therapy of hormonally responsive breast tumors.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/physiology , Receptors, Progesterone/metabolism , Signal Transduction/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , High-Throughput Nucleotide Sequencing , Humans , Promegestone/pharmacology , Promoter Regions, Genetic/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects
17.
PLoS One ; 9(5): e97311, 2014.
Article in English | MEDLINE | ID: mdl-24859236

ABSTRACT

Although non-genomic steroid receptor pathways have been studied over the past decade, little is known about the direct gene expression changes that take place as a consequence of their activation. Progesterone controls proliferation of rat endometrial stromal cells during the peri-implantation phase of pregnancy. We showed that picomolar concentration of progestin R5020 mimics this control in UIII endometrial stromal cells via ERK1-2 and AKT activation mediated by interaction of Progesterone Receptor (PR) with Estrogen Receptor beta (ERb) and without transcriptional activity of endogenous PR and ER. Here we identify early downstream targets of cytoplasmic PR signaling and their possible role in endometrial stromal cell proliferation. Microarray analysis of global gene expression changes in UIII cells treated for 45 min with progestin identified 97 up- and 341 down-regulated genes. The most over-represented molecular functions were transcription factors and regulatory factors associated with cell proliferation and cell cycle, a large fraction of which were repressors down-regulated by hormone. Further analysis verified that progestins regulate Ccnd1, JunD, Usf1, Gfi1, Cyr61, and Cdkn1b through PR-mediated activation of ligand-free ER, ERK1-2 or AKT, in the absence of genomic PR binding. ChIP experiments show that progestin promoted the interaction of USF1 with the proximal promoter of the Cdc2 gene. Usf1 knockdown abolished Cdc2 progestin-dependent transcriptional regulation and cell proliferation, which also blocked Cdc2 knockdown. We conclude that progestin-induced proliferation of endometrial stromal cells is mediated by ERK1-2 and AKT dependent early regulation of USF1, which directly induces Cdc2. To our knowledge, this is the first description of early target genes of progestin-activated classical PR via crosstalk with protein kinases and independently of hormone receptor binding to the genomic targets.


Subject(s)
CDC2 Protein Kinase/metabolism , Chromatin/metabolism , Endometrium/cytology , Gene Expression Regulation/drug effects , Progestins/pharmacology , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Animals , CREB-Binding Protein/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Regulatory Networks/drug effects , Humans , Promegestone/pharmacology , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Rats , Stromal Cells/cytology , Stromal Cells/drug effects , Transcription Factors/metabolism , Upstream Stimulatory Factors/metabolism
18.
Oncogene ; 33(17): 2215-24, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-23708665

ABSTRACT

Prolactin controls the development and function of milk-producing breast epithelia but also supports growth and differentiation of breast cancer, especially luminal subtypes. A principal signaling mediator of prolactin, Stat5, promotes cellular differentiation of breast cancer cells in vitro, and loss of active Stat5 in tumors is associated with antiestrogen therapy failure in patients. In luminal breast cancer, progesterone induces a cytokeratin-5 (CK5)-positive basal cell-like population. This population possesses characteristics of tumor stem cells including quiescence, therapy resistance and tumor-initiating capacity. Here we report that prolactin counteracts induction of the CK5-positive population by the synthetic progestin (Pg) R5020 in luminal breast cancer cells both in vitro and in vivo. CK5-positive cells were chemoresistant as determined by fourfold reduced rate of apoptosis following docetaxel exposure. Pg-induction of CK5 was preceded by marked upregulation of BCL6, an oncogene and transcriptional repressor critical for the maintenance of leukemia-initiating cells. Knockdown of BCL6 prevented induction of CK5-positive cell population by Pg. Prolactin suppressed Pg-induced BCL6 through Jak2-Stat5 but not Erk- or Akt-dependent pathways. In premenopausal but not postmenopausal patients with hormone receptor-positive breast cancer, tumor protein levels of CK5 correlated positively with BCL6, and high BCL6 or CK5 protein levels were associated with unfavorable clinical outcome. Suppression of Pg-induction of CK5-positive cells represents a novel prodifferentiation effect of prolactin in breast cancer. The present progress may have direct implications for breast cancer progression and therapy as loss of prolactin receptor-Stat5 signaling occurs frequently and BCL6 inhibitors currently being evaluated for lymphomas may have value for breast cancer.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Keratin-5/metabolism , Prolactin/physiology , Animals , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Gene Expression , Humans , Keratin-5/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/mortality , Neoplasms, Hormone-Dependent/pathology , Premenopause , Progesterone/physiology , Progesterone Congeners/pharmacology , Promegestone/pharmacology , Proto-Oncogene Proteins c-bcl-6 , Receptors, Estrogen/metabolism , STAT5 Transcription Factor/metabolism
19.
Tissue Eng Part C Methods ; 20(1): 42-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23675751

ABSTRACT

The establishment of hormone target breast cells in the 1970's resulted in suitable models for the study of hormone control of cell proliferation and gene expression using two-dimensional (2D) cultures. However, to study mammogenesis and breast tumor development in vitro, cells must be able to organize in three-dimensional (3D) structures like in the tissue. We now report the development of a hormone-sensitive 3D culture model for the study of mammogenesis and neoplastic development. Hormone-sensitive T47D breast cancer cells respond to estradiol in a dose-dependent manner by forming complex epithelial structures. Treatment with the synthetic progestagen promegestone, in the presence of estradiol, results in flat epithelial structures that display cytoplasmic projections, a phenomenon reported to precede side-branching. Additionally, as in the mammary gland, treatment with prolactin in the presence of estradiol induces budding structures. These changes in epithelial organization are accompanied by collagen remodeling. Collagen is the major acellular component of the breast stroma and an important player in tumor development and progression. Quantitative analysis of second harmonic generation of collagen fibers revealed that collagen density was more variable surrounding budding and irregularly shaped structures when compared to more regular structures; suggesting that fiber organization in the former is more anisotropic than in the latter. In sum, this new 3D model recapitulates morphogenetic events modulated by mammogenic hormones in the breast, and is suitable for the evaluation of therapeutic agents.


Subject(s)
Epithelium/growth & development , Estradiol/pharmacology , Mammary Glands, Human/growth & development , Models, Biological , Promegestone/pharmacology , Tissue Culture Techniques/methods , Actins/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen/metabolism , Epithelium/drug effects , Estrogen Receptor alpha/metabolism , Humans , Mammary Glands, Human/drug effects , Rats , Receptors, Progesterone/metabolism
20.
Mol Hum Reprod ; 20(3): 222-34, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24219889

ABSTRACT

Previous microarray analyses indicated that a portion of the transcriptome in the macaque corpus luteum (CL) of the menstrual cycle was regulated indirectly by luteinizing hormone via the local actions of steroid hormones, notably progesterone (P). The current study was designed to investigate this concept in the CL of early pregnancy by analyzing chorionic gonadotrophin (CG)-regulated genes that are dependent versus independent of local steroid action. Exogenous human chorionic gonadotropin treatment simulating early pregnancy (SEP) began on Day 9 of the luteal phase in female rhesus monkeys with and without concurrent administration of the 3-ß-hydroxysteroid dehydrogenase inhibitor trilostane (TRL) with or without the synthetic progestin R5020. Compared with SEP treatment alone, TRL altered 50 mRNA transcripts on Day 10, rising to 95 on Day 15 (P<0.05, ≥2-fold change in gene expression). Steroid-sensitive genes were validated; notably effects of steroid ablation and P replacement varied by day. Expression of some genes previously identified as P-regulated in the macaque CL during the menstrual cycle were not significantly altered by steroid ablation and P replacement during CG exposure in SEP. These data indicate that the majority of CG-regulated luteal transcripts are differentially expressed independently of local steroid actions. However, the steroid-regulated genes in the macaque CL may be essential during early pregnancy, based on previous reports that TRL treatment initiates premature structural regression of the CL during SEP. These data reinforce the concept that the structure, function and regulation of the rescued CL in early pregnancy differs from the CL of the menstrual cycle in primates.


Subject(s)
Abortifacient Agents, Steroidal/pharmacology , Corpus Luteum/drug effects , Dihydrotestosterone/analogs & derivatives , Progesterone Congeners/pharmacology , Promegestone/pharmacology , RNA, Messenger/genetics , Transcriptome/drug effects , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Chorionic Gonadotropin/pharmacology , Corpus Luteum/metabolism , Dihydrotestosterone/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Luteal Phase/drug effects , Luteal Phase/genetics , Luteinizing Hormone/pharmacology , Macaca mulatta , Pregnancy , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...