Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Endocrinol Metab ; 98(12): E1979-87, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24037887

ABSTRACT

CONTEXT: Peutz-Jeghers syndrome (PJS) is an autosomal-dominant disorder that arises as a consequence of mutations in the STK11 gene that encodes LKB1. PJS males often have estrogen excess manifesting as gynecomastia and advanced bone age. We and others have previously described an increase in testicular aromatase expression in PJS patients. However, the underlying mechanism has not yet been explored. OBJECTIVE: The aim of this study was to characterize the role of LKB1 in regulating the expression of aromatase in boys with PJS via signaling pathways involving AMP-activated protein kinase (AMPK) and cyclic AMP-responsive element binding protein-regulated transcription coactivators (CRTCs). PATIENTS: We studied testicular biopsies from two boys with STK11 mutations: a 13-year-old boy and an unrelated 4-year-old boy with prepubertal gynecomastia and advanced bone age, as well as breast tissue from the 13-year-old boy. RESULTS: Loss of heterozygosity of STK11, measured by the absence of LKB1 immunofluorescence, was observed in Sertoli cells of abnormal cords of testis samples from affected individuals. This was associated with loss of p21 expression and decreased phosphorylation of AMPK, known downstream targets of LKB1, as well as the increased expression of aromatase. Similar results of low LKB1 expression in cells expressing aromatase were observed in the mammary epithelium from one of these individuals. Nuclear expression of the CRTC proteins, potent stimulators of aromatase and known to be inhibited by AMPK, was significantly correlated with aromatase. CONCLUSIONS: Loss of heterozygosity of the STK11 gene leads to an increase in aromatase expression associated with an increase in CRTC nuclear localization, thereby providing a mechanism whereby PJS results in increased endogenous estrogens in affected males.


Subject(s)
Aromatase/biosynthesis , Gynecomastia/etiology , Loss of Heterozygosity , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , Testis/enzymology , AMP-Activated Protein Kinase Kinases , Adolescent , Cell Nucleus/metabolism , Cell Nucleus/pathology , Child, Preschool , Humans , Male , Mammary Glands, Human/enzymology , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Peutz-Jeghers Syndrome/metabolism , Peutz-Jeghers Syndrome/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Sertoli Cells/enzymology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Testis/metabolism , Testis/pathology , Transcription Factors/metabolism
2.
Breast Cancer Res ; 15(2): R30, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23566437

ABSTRACT

INTRODUCTION: The majority of postmenopausal breast cancers are estrogen-dependent. Tumor-derived factors, such as prostaglandin E2 (PGE2), stimulate CREB1 binding to cAMP response elements (CREs) on aromatase promoter II (PII), leading to the increased expression of aromatase and biosynthesis of estrogens within human breast adipose stromal cells (ASCs). Hypoxia inducible factor-1α (HIF-1α), a key mediator of cellular adaptation to low oxygen levels, is emerging as a novel prognostic marker in breast cancer. We have identified the presence of a consensus HIF-1α binding motif overlapping with the proximal CRE of aromatase PII. However, the regulation of aromatase expression by HIF-1α in breast cancer has not been characterized. This study aimed to characterize the role of HIF-1α in the activation of aromatase PII. METHODS: HIF-1α expression and localization were examined in human breast ASCs using quantitative PCR (QPCR), Western blotting, immunofluorescence and high content screening. QPCR and tritiated water-release assays were performed to assess the effect of HIF-1α on aromatase expression and activity. Reporter assays and chromatin immunoprecipitation (ChIP) were performed to assess the effect of HIF-1α on PII activity and binding. Treatments included PGE2 or DMOG ((dimethyloxalglycine), HIF-1α stabilizer). Double immunohistochemistry for HIF-1α and aromatase was performed on tissues obtained from breast cancer and cancer-free patients. RESULTS: Results indicate that PGE2 increases HIF-1α transcript and protein expression, nuclear localization and binding to aromatase PII in human breast ASCs. Results also demonstrate that HIF-1α significantly increases PII activity, and aromatase transcript expression and activity, in the presence of DMOG and/or PGE2, and that HIF-1α and CREB1 act co-operatively on PII. There is a significant increase in HIF-1α positive ASCs in breast cancer patients compared to cancer-free women, and a positive association between HIF-1α and aromatase expression. CONCLUSIONS: This study is the first to identify HIF-1α as a modulator of PII-driven aromatase expression in human breast tumor-associated stroma and provides a novel mechanism for estrogen regulation in obesity-related, post-menopausal breast cancer. Together with our on-going studies on the role of AMP-activated protein kinase (AMPK) in the regulation of breast aromatase, this work provides another link between disregulated metabolism and breast cancer.


Subject(s)
Adipose Tissue/metabolism , Aromatase/genetics , Breast Neoplasms/metabolism , Dinoprostone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Stromal Cells/metabolism , Adipose Tissue/drug effects , Adipose Tissue/pathology , Aromatase/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Female , Fluorescent Antibody Technique , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Oxytocics/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Cells, Cultured
3.
Horm Cancer ; 4(4): 233-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23584792

ABSTRACT

The dramatically increased prevalence of breast cancer after menopause is of great concern and is correlated with elevated local levels of estrogens. This is mainly due to an increase in aromatase expression driven by its proximal promoter II (PII). We have previously demonstrated that the CREB co-activator CRTC2 binds directly to PII and stimulates its activity via mechanisms involving LKB1-AMPK in response to prostaglandin E(2) (PGE(2)). There are three members of the CRTC family (CRTC1-3) and this study aimed to characterize the role of other CRTCs in the activation of aromatase PII. The expression and subcellular localization of CRTCs were examined in preadipocytes using qPCR and immunofluorescence. Under basal conditions, CRTC1 expression was the lowest, whereas CRTC3 transcripts were present at higher levels. Basally, CRTC2 and CRTC3 were mainly cytoplasmic and PGE(2) caused their nuclear translocation. Reporter assays and chromatin immunoprecipitation (ChIP) were performed to assess the effect of CRTCs on PII activity and binding. Basal PII activity was significantly increased with all CRTCs. Forskolin (FSK)/phorbol 12-myristate 13-acetate (PMA), to mimic PGE(2), resulted in a further significant increase in PII activity with all CRTCs, with CRTC2 and CRTC3 having greater effects. This was consistent with ChIP data showing an increased binding of CRTCs to PII with FSK/PMA. Moreover, gene silencing of CRTC2 and CRTC3 significantly reduced the FSK/PMA-mediated stimulation of aromatase activity. Interestingly, CRTCs acted cooperatively with CREB1 to increase PII activity, and both CREs were found to be essential for the maximal induction of PII activity by CRTCs. Phosphorylation of CRTC2 at its AMPK target site, Ser 171, dictated its subcellular localization, and the activation of aromatase PII in preadipocytes. In conclusion, this study demonstrates that aromatase regulation in primary human breast preadipocytes involves more than one CRTC.


Subject(s)
Adipocytes/enzymology , Aromatase/genetics , Transcription Factors/physiology , 3T3 Cells , Animals , Aromatase/metabolism , COS Cells , Chlorocebus aethiops , Cyclic AMP Response Element-Binding Protein/metabolism , Dinoprostone/physiology , Enzyme Induction , Humans , Mice , Phosphorylation , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Transcription Factors/metabolism , Transcription, Genetic
4.
Mol Cell Endocrinol ; 366(2): 170-9, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-22801104

ABSTRACT

AMP-activated protein kinase (AMPK) is a master regulator of energy homeostasis involved in the regulation of a number of physiological processes including ß-oxidation of fatty acids, lipogenesis, protein and cholesterol synthesis, as well as cell cycle inhibition and apoptosis. Important changes to these processes are known to occur in cancer due to changes in AMPK activity within cancer cells and in the periphery. This review aims to present findings relating to the role and regulation of AMPK in endocrine-related cancers. Obesity is a known risk factor for many types of cancers and a number of endocrine factors, including adipokines and steroid hormones, are regulated by and regulate AMPK. A clear role for AMPK in breast cancer is evident from the already impressive body of work published to date. However, information pertaining to its role in prostate cancer is still contentious, and future work should unravel the intricacies behind its role to inhibit, in some cases, and stimulate cancer growth in others. This review also presents data relating to the role of AMPK in cancers of the endometrium, ovary and colon, and discusses the possible use of AMPK-activating drugs including metformin for the treatment of all endocrine-related cancers.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Colonic Neoplasms/enzymology , Endometrial Neoplasms/enzymology , Obesity/enzymology , Ovarian Neoplasms/enzymology , Prostatic Neoplasms/enzymology , AMP-Activated Protein Kinases/genetics , Biological Factors/pharmacology , Cell Transformation, Neoplastic , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Endocrine System/drug effects , Endocrine System/enzymology , Endocrine System/pathology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Metformin/pharmacology , Metformin/therapeutic use , Obesity/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics
5.
Steroids ; 76(8): 768-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21414336

ABSTRACT

Phase III aromatase inhibitors (AIs) are proving successful in the treatment of hormone-dependent postmenopausal breast cancer. Side-effects associated with total body aromatase inhibition have prompted new research into the development of breast-specific AIs. The identification of tissue- and disease-specific usage of aromatase promoters has made the inhibition of aromatase at the transcriptional level an interesting approach. We have previously demonstrated that AMPK-activating drugs, including metformin, were potent inhibitors of aromatase expression in primary human breast adipose stromal cells (hASCs). This study examines the promoter-specific effects of metformin on inhibiting aromatase expression in hASCs. Tumour-associated promoters PII/PI.3 were activated using forskolin (FSK)/phorbol ester (PMA), whereas normal adipose associated promoter PI.4 was activated using dexamethasone (DEX)/tumour necrosis factor-α (TNFα). Results demonstrate that metformin significantly decreased the FSK/PMA-, but not the DEX/TNFα-mediated expression of total aromatase at concentrations of 10, 20, and 50 µM (P ≤ 0.05). Using PCR to amplify promoter-specific transcripts of aromatase, it appears that the inhibition of the FSK/PMA-mediated expression of aromatase is due to decreases in PII/PI.3-specific transcripts, whereas no effect of metformin is observed on any promoter-specific transcript, including PI.4, in DEX/TNFα-treated hASCs. This report therefore supports the hypothesis that metformin would act as a breast-specific inhibitor of aromatase expression in the context of postmenopausal breast cancer.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/genetics , Breast/enzymology , Metformin/pharmacology , Promoter Regions, Genetic/drug effects , Adipose Tissue/enzymology , Cells, Cultured , Colforsin/pharmacology , Dexamethasone/pharmacology , Female , Gene Expression Regulation , Humans , Postmenopause , Stromal Cells/enzymology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...