Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Hum Mutat ; 39(12): 1861-1874, 2018 12.
Article in English | MEDLINE | ID: mdl-30067310

ABSTRACT

Nuclear receptor subfamily 5 group A member 1/Steroidogenic factor 1 (NR5A1; SF-1; Ad4BP) mutations cause 46,XY disorders of sex development (DSD), with phenotypes ranging from developmentally mild (e.g., hypospadias) to severe (e.g., complete gonadal dysgenesis). The molecular mechanism underlying this spectrum is unclear. During sex determination, SF-1 regulates SOX9 (SRY [sex determining region Y]-box 9) expression. We hypothesized that SF-1 mutations in 46,XY DSD patients affect SOX9 expression via the Testis-specific Enhancer of Sox9 core element, TESCO. Our objective was to assess the ability of 20 SF-1 mutants found in 46,XY DSD patients to activate TESCO. Patient DNA was sequenced for SF-1 mutations and mutant SF-1 proteins were examined for transcriptional activity, protein expression, sub-cellular localization and in silico structural defects. Fifteen of the 20 mutants showed reduced SF-1 activation on TESCO, 11 with atypical sub-cellular localization. Fourteen SF-1 mutants were predicted in silico to alter DNA, ligand or cofactor interactions. Our study may implicate aberrant SF-1-mediated transcriptional regulation of SOX9 in 46,XY DSDs.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Enhancer Elements, Genetic , Mutation , SOX9 Transcription Factor/genetics , Steroidogenic Factor 1/genetics , Adolescent , Adult , Child , Child, Preschool , Computer Simulation , Gene Expression Regulation , HEK293 Cells , Humans , Infant , Infant, Newborn , Ligands , Male , Protein Binding , Sequence Analysis, DNA/methods , Steroidogenic Factor 1/chemistry , Steroidogenic Factor 1/metabolism
2.
J Mol Biol ; 430(10): 1531-1543, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29555554

ABSTRACT

Activation of estrogen receptor α (ERα) stimulates cell division and tumor growth by modulating the expression of ERα target genes. This activation involves the recruitment of specific proteins with activities that are still not fully understood. Timeless, the human homolog of the Drosophila gene involved in circadian rhythm, was previously shown to be a strong predictor of tamoxifen relapse, and is involved in genomic stability and cell cycle control. In this study, we investigated the interplay between Timeless and ERα, and showed that human Timeless is an ERα coactivator. Timeless binds to ERα and enhances its transcriptional activity. Overexpressing Timeless increases PARP1 expression and enhances ERα-induced gene regulation through the proximal LXXLL motif on Timeless protein and ERα PARylation. Finally, Timeless is recruited with ERα on the GREB1 and cMyc promoters. These data, the first to link Timeless to steroid hormone function, provide a mechanistic basis for its clinical association with tamoxifen resistance. Thus, our results identify Timeless as another key regulator of ERα in controlling ERα transactivation.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Estrogen Receptor alpha/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Transcriptional Activation , Breast Neoplasms/metabolism , Cell Proliferation , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Prognosis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Survival Analysis
3.
J Steroid Biochem Mol Biol ; 145: 179-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24846828

ABSTRACT

Aromatase is the critical enzyme that converts androgens to estrogens. It is frequently highly expressed in the tumour bearing breast of women diagnosed with estrogen receptor positive tumours, resulting in dramatically increased local estrogen production to drive tumour progression. Expression of aromatase is regulated primarily at the transcriptional level of its encoding gene CYP19A1, located on chromosome 15 of the human genome. A characteristic feature of CYP19A1 expression is its use of alternative promoters to regulate transcription in a tissue-specific manner. In breast cancer, the increase in aromatase expression is mediated via higher expression of the distal adipose-specific promoter I.4 and a switch to the preferential use of proximal promoters I.3 and II. This results in a net increase of CYP19A1 transcripts in tumour-bearing breast up to 3-4-fold higher than normal breast. Current aromatase inhibitors - whilst efficacious - exhibit significant side effects that reduce patient compliance. Understanding the transcription factors and signalling pathways that control aromatase expression will lead to opportunities to develop breast-specific inhibitors with an improved side-effects profile. This article is part of a Special Issue entitled 'Essential role of DHEA'.


Subject(s)
Aromatase/metabolism , Breast/metabolism , Estrogens/biosynthesis , Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Receptors, Estrogen/metabolism , Transcription, Genetic , Animals , Aromatase/genetics , Breast Neoplasms/metabolism , Female , Humans , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal , Mice , Mice, Knockout , Promoter Regions, Genetic
4.
Mol Cell Endocrinol ; 394(1-2): 21-8, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25004254

ABSTRACT

The cytokine Tumor Necrosis Factor-α is critical to Estrogen Receptor positive breast cancer pathology, stimulating estrogen-biosynthesis pathways and preventing the differentiation of estrogen-producing fibroblasts. High concentrations of TNFα are detected in the tumor microenvironment, and infiltrating immune cells are thought to be a major source. This study identifies that TNFα is also a tumor-derived factor, expressed in ER+ tumour epithelial cells and regulated by 17-ß-estradiol (E2). Treatment of MCF-7, T47D and ZR-75 breast cancer cells with E2 increased TNFα mRNA and protein expression and secretion. This effect was mitigated with the use of ERα inhibitors 4-hydroy-tamoxifen and ICI-182780, indicating that E2-mediated TNFα induction was via the actions of ERα. Chromatin immunoprecipitation reveals ERα binding to the TNFα promoter upon stimulation with E2. This study demonstrates for the first time a positive feedback loop between estradiol and TNFα, critical in maintaining high levels of the hormone within the ER+ breast tumour microenvironment.


Subject(s)
Epithelial Cells/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Feedback, Physiological , Female , Fulvestrant , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Signal Transduction , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/metabolism
5.
Endocr Relat Cancer ; 21(2): T33-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24532474

ABSTRACT

The heritable component of breast cancer accounts for only a small proportion of total incidences. Environmental and lifestyle factors are therefore considered to among the major influencing components increasing breast cancer risk. Endocrine-disrupting chemicals (EDCs) are ubiquitous in the environment. The estrogenic property of EDCs has thus shown many associations between ongoing exposures and the development of endocrine-related diseases, including breast cancer. The environment consists of a heterogenous population of EDCs and despite many identified modes of action, including that of altering the epigenome, drawing definitive correlations regarding breast cancer has been a point of much discussion. In this review, we describe in detail well-characterized EDCs and their actions in the environment, their ability to disrupt mammary gland formation in animal and human experimental models and their associations with exposure and breast cancer risk. We also highlight the susceptibility of early-life exposure to each EDC to mediate epigenetic alterations, and where possible describe how these epigenome changes influence breast cancer risk.


Subject(s)
Breast Neoplasms/etiology , Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Animals , Epigenesis, Genetic , Female , Genome , Humans , Life Style , Phytoestrogens/pharmacology
6.
PLoS One ; 9(2): e87230, 2014.
Article in English | MEDLINE | ID: mdl-24520329

ABSTRACT

Estrogens are known to play a role in modulating metabolic processes within the body. The Aromatase knockout (ArKO) mice have been shown to harbor factors of Metabolic syndrome with central adiposity, hyperinsulinemia and male-specific hepatic steatosis. To determine the effects of estrogen ablation and subsequent replacement in males on whole body glucose metabolism, three- and six-month-old male ArKO mice were subjected to whole body glucose, insulin and pyruvate tolerance tests and analyzed for ensuing metabolic changes in liver, adipose tissue, and skeletal muscle. Estrogen-deficient male ArKO mice showed increased gonadal adiposity which was significantly reduced upon 17ß-estradiol (E2) treatment. Concurrently, elevated ArKO serum leptin levels were significantly reduced upon E2 treatment and lowered serum adiponectin levels were restored to wild type levels. Three-month-old male ArKO mice were hyperglycemic, and both glucose and pyruvate intolerant. These phenotypes continued through to 6 months of age, highlighting a loss of glycemic control. ArKO livers displayed changes in gluconeogenic enzyme expression, and in insulin signaling pathways upon E2 treatment. Liver triglycerides were increased in the ArKO males only after 6 months of age, which could be reversed by E2 treatment. No differences were observed in insulin-stimulated ex vivo muscle glucose uptake nor changes in ArKO adipose tissue and muscle insulin signaling pathways. Therefore, we conclude that male ArKO mice develop hepatic glucose intolerance by the age of 3 months which precedes the sex-specific development of hepatic steatosis. This can be reversed upon the administration of exogenous E2.


Subject(s)
Aromatase/deficiency , Aromatase/metabolism , Glucose Intolerance/enzymology , Liver/metabolism , Liver/pathology , Adiponectin/blood , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Estrogens/pharmacology , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Glucose Intolerance/blood , Glucose Intolerance/pathology , Insulin/blood , Insulin Resistance , Leptin/blood , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscles/metabolism , Organ Size/drug effects , Phosphorylation/drug effects , Pyruvic Acid/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Triglycerides/metabolism
8.
Breast Cancer Res Treat ; 142(1): 211-23, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24122391

ABSTRACT

The interaction between breast tumor epithelial and stromal cells is vital for initial and recurrent tumor growth. While breast cancer-associated stromal cells provide a favorable environment for proliferation and metastasis, the molecular mechanisms contributing to this process are not fully understood. Nuclear receptors (NRs) are intracellular transcription factors that directly regulate gene expression. Little is known about the status of NRs in cancer-associated stroma. Nuclear Receptor Low-Density Taqman Arrays were used to compare the gene expression profiles of all 48 NR family members in a collection of primary cultured cancer-associated fibroblasts (CAFs) obtained from estrogen receptor (ER)α positive breast cancers (n = 9) and normal breast adipose fibroblasts (NAFs) (n = 7). Thirty-three of 48 NRs were expressed in both the groups, while 11 NRs were not detected in either. Three NRs (dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1 (DAX-1); estrogen-related receptor beta (ERR-ß); and RAR-related orphan receptor beta (ROR-ß)) were only detected in NAFs, while one NR (liver receptor homolog-1 (LRH-1)) was unique to CAFs. Of the NRs co-expressed, four were significantly down-regulated in CAFs compared with NAFs (RAR-related orphan receptor-α (ROR-α); Thyroid hormone receptor-ß (TR-ß); vitamin D receptor (VDR); and peroxisome proliferator-activated receptor-γ (PPAR-γ)). Quantitative immunohistochemistry for LRH-1, TR-ß, and PPAR-γ proteins in stromal fibroblasts from an independent panel of breast cancers (ER-positive (n = 15), ER-negative (n = 15), normal (n = 14)) positively correlated with mRNA expression profiles. The differentially expressed NRs identified in tumor stroma are key mediators in aromatase regulation and subsequent estrogen production. Our findings reveal a distinct pattern of NR expression that therefore fits with a sustained and increased local estrogen microenvironment in ER-positive tumors. NRs in CAFs may provide a new avenue for the development of intratumoral-targeted therapies in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Stromal Cells/metabolism , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Receptor, ErbB-2/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Risk Factors
9.
Biochem Biophys Res Commun ; 438(3): 533-9, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23911789

ABSTRACT

The expression of orphan nuclear receptor Liver Receptor Homolog-1 (LRH-1) is elevated in breast cancer and promotes proliferation, migration and invasion in vitro. LRH-1 expression is regulated by oestrogen (E2), with LRH-1 mRNA transcript levels higher in oestrogen receptor α (ERα) positive (ER+) breast cancer cells compared to ER- cells. However, the presence of LRH-1 protein in ER- cells suggests discordance between mRNA transcript levels and protein expression. To understand this, we investigated the impact of mRNA and protein stability in determining LRH-1 protein levels in breast cancer cells. LRH-1 transcript levels were significantly higher in ER+ versus ER- breast cancer cells lines; however LRH-1 protein was expressed at similar levels. We found LRH-1 mRNA and protein was more stable in ER- compared to ER+ cell lines. The tumor-specific LRH-1 variant isoform, LRH-1v4, which is highly responsive to E2, showed increased mRNA stability in ER- versus ER+ cells. In addition, in MCF-7 and T47-D cell lines, LRH-1 total mRNA stability was reduced with E2 treatment, this effect mediated by ERα. Our data demonstrates that in ER- cells, increased mRNA and protein stability contribute to the abundant protein expression levels. Expression and immunolocalisation of LRH-1 in ER- cells as well as ER- tumors suggests a possible role in the development of ER- tumors. The modulation of LRH-1 bioactivity may therefore be beneficial as a treatment option in both ER- and ER+ breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Protein Stability/drug effects , RNA Stability/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Cell Line, Tumor , Female , Humans , Protein Isoforms/genetics , RNA Stability/genetics , RNA, Messenger/metabolism
10.
J Interferon Cytokine Res ; 33(7): 335-45, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23472660

ABSTRACT

Tumor necrosis factor α (TNFα) has many roles in both physiological and pathological states. Initially thought to cause necrosis of tumors, research has shown that in many tumor types, including breast cancer, TNFα contributes to growth and proliferation. The presence of TNFα-derived from the tumor and infiltrating immune cells-within a breast tumor microenvironment has been correlated with a more aggressive phenotype, and the postmenopausal ER+ subtype of breast cancers appears to strongly respond to its many pro-growth signaling functions. We discuss how TNFα regulates estrogen biosynthesis within the breast, affecting the activity of the key estrogen-synthesizing enzymes aromatase, estrone sulfatase, and 17ß-HSD type 1. Additionally, we describe the anti-adipogenic actions of TNFα that are critical in preventing adjacent estrogen-producing adipose fibroblasts from differentiating, ensuring that the tumor maintains a constant source of estrogen-producing cells. We examine how the increased risk of developing breast cancer in older and obese individuals may be linked to the levels of TNFα in the body. Finally, we evaluate the feasibility of targeting TNFα and its associated pathways as a novel approach to breast cancer therapeutics.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adipogenesis , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/therapy , Carcinogenesis , Female , Humans , Immunotherapy/methods , Molecular Targeted Therapy , Postmenopause , Risk Factors , Tumor Microenvironment , Tumor Necrosis Factor-alpha/antagonists & inhibitors
11.
Biochem Biophys Res Commun ; 433(1): 96-101, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23485457

ABSTRACT

The Early Growth Response genes EGR2 and EGR3 play an important role in mediating TNFα induced aromatase expression via the adipose specific promoter PI.4. The upstream signalling pathway stimulated by TNFα to initiate this is unknown. The aim of this present study was to determine the signalling pathways activated by TNFα which result in EGR2 and EGR3 transcription, and ultimately activation of PI.4. The NFκB inhibitor BAY-11-7082 dose-dependently inhibited transcription of EGR2 and EGR3 mRNA as well as total and PI.4-specific CYP19A1 mRNA. The MAPK pathway inhibitor U0126, inhibitor of MEK1/2 had the same effect, however inhibition of c-Jun and JNK1,2,3 with SP600125 did not lead to down-regulation. We provide evidence for the first time that EGR2 and EGR3 are regulated by NFκB and MAPK signalling pathways downstream of TNFα stimulation in breast adipose fibroblasts, and that this in turn is upstream of CYP19A1 transcription via PI.4.


Subject(s)
Aromatase/genetics , Early Growth Response Protein 2/genetics , Early Growth Response Protein 3/genetics , MAP Kinase Signaling System , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Base Sequence , Breast/cytology , Breast/drug effects , Breast/metabolism , Butadienes/pharmacology , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Humans , MAP Kinase Signaling System/drug effects , Models, Biological , Nitriles/pharmacology , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects
12.
J Steroid Biochem Mol Biol ; 137: 157-64, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23339934

ABSTRACT

Epigenome changes have been widely demonstrated to contribute to the initiation and progression of a vast array of cancers including breast cancer. The reversible process of many epigenetic modifications is thus an attractive feature for the development of novel therapeutic measures. In oestrogen receptor α (hereinafter referred to as ER) positive tumours, endocrine therapies have proven beneficial in patient care, particularly in postmenopausal women where two-thirds of tumours are oestrogen dependent. However, resistance to such therapies is a common feature amongst individuals. In the current review, we discuss the influence that epigenetics has on oestrogen dependent breast cancers, in particular (i) the production of intracrine oestrogen in postmenopausal women, (ii) the action of oestrogen on epigenetic processes, and (iii) the links between epigenetics and endocrine resistance and the current advancements in epigenetic therapy that target this process. This article is part of a Special Issue entitled 'CSR 2013'.


Subject(s)
Breast Neoplasms/physiopathology , Epigenesis, Genetic , Estrogens/biosynthesis , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estrogens/physiology , Female , Humans
13.
Breast Cancer Res Treat ; 138(1): 193-203, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23338760

ABSTRACT

Expression of the oestrogen producing enzyme, aromatase, is regulated in a tissue-specific manner by its encoding gene CYP19A1. In post-menopausal women, the major site for oestrogen production in the breast is the adipose, where CYP19A1 transcription is driven by the distal promoter I.4 (PI.4). Transcripts via this promoter are also elevated in breast adipose fibroblasts (BAFs) adjacent to a tumour. PI.4 expression is stimulated by a number of cytokines, and TNFα is one such factor. The transcriptional mechanisms induced by TNFα to stimulate PI.4 are poorly characterised. We show that the early growth response (Egr) transcription factors play an important role in the TNFα-induced signalling pathway resulting in elevated PI.4 transcription. TNFα treatment of BAFs increases mRNA levels of all four Egr family members, with EGR2 being the most highly expressed. Overexpression of EGR2 causes an increase in endogenous CYP19A1 expression in preadipocyte Simpson-Golabi-Behmel syndrome cells, driven by increases in PI.4-specific transcripts. PI.4 luciferase reporter activity is increased in a dose-dependent manner by EGR2, EGR3 and EGR4, with EGR2 showing the most potent activation of promoter activity. Deletion analysis indicates that this promoter activity is being indirectly mediated by a short region of the promoter not containing any previously characterised binding sites, and we further show that EGR2 does not bind directly or indirectly to this promoter region. However, siRNA knockdown of the Egrs reduces the total and PI.4-derived CYP19A1 transcription in BAFs. These studies unveil a novel component of the aromatase gene regulatory network and further enhance the complexity of oestrogen production in the breast.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/metabolism , Aromatase/genetics , Breast/cytology , Early Growth Response Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Early Growth Response Transcription Factors/genetics , Female , Gene Expression , Genes, Reporter , Humans , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Messenger/genetics , Response Elements , Sequence Deletion , Transcriptional Activation
14.
J Steroid Biochem Mol Biol ; 132(3-5): 331-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22929011

ABSTRACT

The increase in local oestrogen production seen in oestrogen receptor positive (ER+) breast cancers is driven by increased activity of the aromatase enzyme. CYP19A1, the encoding gene for aromatase, is often overexpressed in the oestrogen-producing cells of the breast adipose fibroblasts (BAFs) surrounding an ER+ tumour, and the molecular processes underlying this upregulation is important in the development of breast-specific aromatase inhibitors for breast cancer therapy. Prostaglandin E2 (PGE2), a factor secreted by tumours, is known to stimulate CYP19A1 expression in human BAFs. The hormonal regulation of this process has been examined; however, what is less well understood is the emerging role of epigenetic mechanisms and how they modulate PGE2 signalling. This present study characterises the epigenetic processes underlying expression of the prostanoid receptor EP2 in the context of ER+ breast cancer. Sodium bisulphite sequencing of CpG methylation within the promoter region of EP2 revealed that an inverse correlation existed between methylation levels and relative EP2 expression in breast cancer cell lines MDA-MB-231, MCF7 and MCF10A but not in HS578t and T47D. Inhibition of DNA methylation with 5-aza-2'-deoxycytidine (5aza) and histone deacetylation with Trichostatin A (TSA) resulted in upregulation of EP2 mRNA in all cell lines with varying influences of each epigenetic process observed. Expression of EP2 was detected in human BAFs despite a natively methylated promoter, and this expression was further increased upon 5aza treatment. An examination of 3 triple negative, 3 ductal carcinoma in situ and 3 invasive ductal carcinoma samples revealed that there was no change in EP2 promoter methylation status between normal and cancer associated stroma, despite observed differences in relative mRNA levels. Although EP2 methylation status is inversely correlated to expression levels in established breast cancer cell lines, we could not identify that such a correlation existed in tumour-associated stroma cells.


Subject(s)
Breast Neoplasms/genetics , Epigenesis, Genetic , Receptors, Prostaglandin E, EP2 Subtype/genetics , Adipose Tissue/cytology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , CpG Islands , DNA Methylation , Decitabine , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Promoter Regions, Genetic , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Reference Values , Stromal Cells/metabolism
15.
PLoS One ; 7(2): e31593, 2012.
Article in English | MEDLINE | ID: mdl-22359603

ABSTRACT

BACKGROUND: Liver Receptor Homolog 1 (LRH-1, NR5A2) is an orphan nuclear receptor that is over-expressed in cancers in tissues such as the breast, colon and pancreas. LRH-1 plays important roles in embryonic development, steroidogenesis and cholesterol homeostasis. In tumor cells, LRH-1 induces proliferation and cell cycle progression. High LRH-1 expression is demonstrated in breast cancers, positively correlating with ERα status and aromatase activity. LRH-1 dependent cellular mechanisms in breast cancer epithelial cells are poorly defined. Hence in the present study we investigated the actions of LRH-1 in estrogen receptor α (ERα) positive breast cancer cells. RESULTS: The study aimed to investigate LRH-1 dependent mechanisms that promote breast cancer proliferation. We identified that LRH-1 regulated the expression of Growth Regulation by Estrogen in Breast Cancer 1 (GREB1) in MCF-7 and MDA-MB-231 cells. Over-expression of LRH-1 increased GREB1 mRNA levels while knockdown of LRH-1 reduced its expression. GREB1 is a well characterised ERα target gene, with three estrogen response elements (ERE) located on its promoter. Chromatin immunoprecipitation studies provided evidence of the co-localisation of LRH-1 and ERα at all three EREs. With electrophoretic mobility shift assays, we demonstrated direct binding of LRH-1 to EREs located on GREB1 and Trefoil Factor 1 (TFF1, pS2) promoters. LRH-1 and ERα co-operatively activated transcription of ERE luciferase reporter constructs suggesting an overlap in regulation of target genes in breast cancer cells. Over-expression of LRH-1 resulted in an increase in cell proliferation. This effect was more pronounced with estradiol treatment. In the presence of ICI 182,780, an ERα antagonist, LRH-1 still induced proliferation. CONCLUSIONS: We conclude that in ER-positive breast cancer cells, LRH-1 promotes cell proliferation by enhancing ERα mediated transcription of target genes such as GREB-1. Collectively these findings indicate the importance of LRH-1 in the progression of hormone-dependent breast cancer and implicate LRH-1 as a potential avenue for drug development.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Estrogen Receptor alpha/physiology , Neoplasm Proteins/genetics , Orphan Nuclear Receptors/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Response Elements , Transcription, Genetic
16.
Breast Cancer Res Treat ; 132(2): 765-71, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22237979

ABSTRACT

The main biological active substance secreted by the pineal gland, melatonin (MLT), counteracts the effects of estrogens in breast cancer via exerting a number of its own oncostatic properties. Recent studies of postmenopausal women have identified that the major metabolite of MLT is statistically significantly associated with a lower risk of developing breast cancer. While MLT production decreases with age, breast cancer risk, however, increases with age and obesity. We hypothesize that MLT inhibits estrogen production in breast adipose fibroblasts (BAFs), the main local source of estrogen in breast tumors of postmenopausal women, by inhibiting transcription of the CYP19A1 gene that encodes the key enzyme aromatase. Normal BAFs were cultured from women undergoing breast reduction surgery, while breast cancer-associated fibroblasts (CAFs) were isolated from three women with estrogen receptor (ER) positive invasive ductal carcinomas. MTNR1A and MTNR1B receptor expression and CYP19A1 mRNA expression following MLT treatments were determined by qRT-PCR. BAFs express the G-protein coupled MLT receptors MTNR1A and MTNR1B with elevated levels of MTNR1A found in CAFs. Treatment of BAFs and CAFs with MLT resulted in significant suppression of CYP19A1 transcription and aromatase activity at pharmacological, physiological and sub-physiological concentrations. MLT suppression occurred through promoter-specific PI.4-, PI.3- and PII-derived CYP19A1 mRNA. Stimulation of CYP19A1 PII-mRNA and aromatase activity by prostaglandin E(2) (PGE(2)) were significantly attenuated by physiological doses of MLT. Lower levels of MLT in aging women may increase the risk of progressing ER-positive breast cancer through a decreased ability to suppress CYP19A1 expression and subsequent local estrogen production in BAFs/CAFs.


Subject(s)
Adipose Tissue/enzymology , Aromatase/metabolism , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Fibroblasts/enzymology , Melatonin/metabolism , Adipose Tissue/pathology , Aromatase/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cells, Cultured , Dinoprostone/metabolism , Down-Regulation , Female , Fibroblasts/pathology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Receptors, Estrogen/metabolism , Transcription, Genetic
17.
Chromosome Res ; 20(1): 191-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22215485

ABSTRACT

In most mammals, the Y chromosomal Sry gene initiates testis formation within the bipotential gonad, resulting in male development. SRY is a transcription factor and together with SF1 it directly up-regulates the expression of the pivotal sex-determining gene Sox9 via a 1.3-kb cis-regulatory element (TESCO) which contains an evolutionarily conserved region (ECR) of 180 bp. Remarkably, several rodent species appear to determine sex in the absence of Sry and a Y chromosome, including the mole voles Ellobius lutescens and Ellobius tancrei, whereas Ellobius fuscocapillus of the same genus retained Sry. The sex-determining mechanisms in the Sry-negative species remain elusive. We have cloned and sequenced 1.1 kb of E. lutescens TESCO which shares 75% sequence identity with mouse TESCO indicating that testicular Sox9 expression in E. lutescens might still be regulated via TESCO. We have also cloned and sequenced the ECRs of E. tancrei and E. fuscocapillus. While the three Ellobius ECRs are highly similar (94-97% sequence identity), they all display a 14-bp deletion (Δ14) removing a highly conserved SOX/TCF site. Introducing Δ14 into mouse TESCO increased both basal activity and SF1-mediated activation of TESCO in HEK293T cells. We propose a model whereby Δ14 may have triggered up-regulation of Sox9 in XX gonads leading to destabilization of the XY/XX sex-determining mechanism in Ellobius. E. lutescens/E. tancrei and E. fuscocapillus could have independently stabilized their sex determination mechanisms by Sry-independent and Sry-dependent approaches, respectively.


Subject(s)
Arvicolinae/genetics , Gene Expression Regulation , SOX9 Transcription Factor/metabolism , Sex Determination Processes , Y Chromosome/metabolism , Animals , Arvicolinae/metabolism , Arvicolinae/physiology , Base Sequence , Binding Sites , Cloning, Molecular , Conserved Sequence , Evolution, Molecular , Female , Genetic Variation , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Male , Mice , Models, Genetic , Molecular Sequence Data , Mutagenesis, Site-Directed , SOX9 Transcription Factor/genetics , Sequence Alignment , Sequence Analysis, DNA , Sequence Deletion , Testis/cytology , Testis/metabolism , Testis/physiology , Y Chromosome/genetics
18.
PLoS One ; 6(3): e17751, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21412441

ABSTRACT

BACKGROUND: In human embryogenesis, loss of SRY (sex determining region on Y), SOX9 (SRY-related HMG box 9) or SF1 (steroidogenic factor 1) function causes disorders of sex development (DSD). A defining event of vertebrate sex determination is male-specific upregulation and maintenance of SOX9 expression in gonadal pre-Sertoli cells, which is preceded by transient SRY expression in mammals. In mice, Sox9 regulation is under the transcriptional control of SRY, SF1 and SOX9 via a conserved testis-specific enhancer of Sox9 (TES). Regulation of SOX9 in human sex determination is however poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: We show that a human embryonal carcinoma cell line (NT2/D1) can model events in presumptive Sertoli cells that initiate human sex determination. SRY associates with transcriptionally active chromatin in NT2/D1 cells and over-expression increases endogenous SOX9 expression. SRY and SF1 co-operate to activate the human SOX9 homologous TES (hTES), a process dependent on phosphorylated SF1. SOX9 also activates hTES, augmented by SF1, suggesting a mechanism for maintenance of SOX9 expression by auto-regulation. Analysis of mutant SRY, SF1 and SOX9 proteins encoded by thirteen separate 46,XY DSD gonadal dysgenesis individuals reveals a reduced ability to activate hTES. CONCLUSIONS/SIGNIFICANCE: We demonstrate how three human sex-determining factors are likely to function during gonadal development around SOX9 as a hub gene, with different genetic causes of 46,XY DSD due a common failure to upregulate SOX9 transcription.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Mutation/genetics , SOX9 Transcription Factor/genetics , Sex-Determining Region Y Protein/genetics , Steroidogenic Factor 1/genetics , Cell Line , Enhancer Elements, Genetic/genetics , Humans , Male , Mutant Proteins/metabolism , Organ Specificity/genetics , SOX9 Transcription Factor/metabolism , Testis/metabolism , Trans-Activators/metabolism , Up-Regulation/genetics
19.
PLoS One ; 5(12): e14389, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-21188138

ABSTRACT

BACKGROUND: Granulosa cell tumors (GCT) of the ovary often express aromatase and synthesize estrogen, which in turn may influence their progression. Recently a specific point mutation (C134W) in the FOXL2 protein was identified in >94% of adult-type GCT and it is likely to contribute to their development. A number of genes are known to be regulated by FOXL2, including aromatase/CYP19A1, but it is unclear which are direct targets and whether the C134W mutation alters their regulation. Recently, it has been reported that FOXL2 forms a complex with steroidogenic factor 1 (SF-1) which is a known regulator of aromatase in granulosa cells. METHODOLOGY/PRINCIPAL FINDINGS: In this work, the human GCT-derived cell lines KGN and COV434 were heterozygous and wildtype for the FOXL2:C134W mutation, respectively. KGN had abundant FOXL2 mRNA expression but it was not expressed in COV434. Expression of exogenous FOXL2:C134W in COV434 cells induced higher expression of a luciferase reporter for the ovarian specific aromatase promoter, promoter II (PII) (-516bp) than expression of wildtype FOXL2, but did not alter induction of a similar reporter for the steroidogenic acute regulatory protein (StAR) promoter (-1300bp). Co-immunoprecipitation confirmed that FOXL2 bound SF-1 and that it also bound its homologue, liver receptor homologue 1 (LRH-1), however, the C134W mutation did not alter these interactions or induce a selective binding of the proteins. A highly conserved putative binding site for FOXL2 was identified in PII. FOXL2 was demonstrated to bind the site by electrophoretic mobility shift assays (EMSA) and site-directed mutagenesis of this element blocked its differential induction by wildtype FOXL2 and FOXL2:C134W. CONCLUSIONS/SIGNIFICANCE: These findings suggest that aromatase is a direct target of FOXL2:C134W in adult-type GCT via a single distinctive and highly conserved binding site in PII and therefore provide insight into the pathogenic mechanism of this mutation.


Subject(s)
Aromatase/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Granulosa Cell Tumor/metabolism , Mutation , Ovarian Neoplasms/genetics , Promoter Regions, Genetic , Binding Sites , Cell Line, Tumor , Female , Forkhead Box Protein L2 , Humans , Mutagenesis, Site-Directed , Phosphoproteins/genetics , Point Mutation , Regulatory Sequences, Nucleic Acid
20.
Mol Cell Endocrinol ; 321(2): 123-30, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20211687

ABSTRACT

Cytochrome aromatase p450, encoded by the gene CYP19, catalyzes the synthesis of estrogens from androgens. In post-menopausal women, adipose becomes the major site for estrogen production, where basal CYP19 transcription is driven by distal promoter I.4. In breast adipose fibroblasts (BAFs), CYP19 expression is elevated in the presence of tumour-derived factors through use of promoters I.3 and II. We show for the first time that DNA methylation contributes to CYP19 regulation in BAFs and breast cell lines. Promoter I.4 and I.3/II-derived mRNA were not dependent on the CpG methylation status within respective promoters. However, inhibition of DNA methylation with 5-aza-2'-deoxycytidine resulted in a significant approximately 40-fold induction in CYP19 mRNA expression in BAFs and breast cell lines. These studies uncover a new layer of complexity in the regulation of aromatase where CYP19 appears to be inhibited by DNA methylation and evokes the possibility that disruption to this epigenetic regulation may give rise to an increase in aromatase levels in the breast.


Subject(s)
Aromatase/metabolism , Breast Neoplasms/metabolism , Epigenesis, Genetic , Fibroblasts/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , Adipose Tissue/cytology , Adipose Tissue/metabolism , Aromatase/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , CpG Islands/genetics , DNA Methylation/physiology , DNA-Cytosine Methylases/metabolism , Female , Humans , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...