1
|
Thamnawasolos J, Boonphakdee C. Cyp19 expression and sex change timing in captive-bred false clownfish. BRAZ J BIOL 2023; 83:e273824. [PMID: 37585931 DOI: 10.1590/1519-6984.273824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/20/2023] [Indexed: 08/18/2023] Open
Abstract
The false clown anemonefish (Amphiprion ocellaris) is a protandrous hermaphrodite with a distinctive reproductive behavior. This study elucidates the genetic mechanisms and timing of sex changes in captive-bred A. ocellaris by examining the expression of key genes involved in this process, specifically cyp19a1a and cyp19a1b. Gonadal histological analyses and gene expression studies were conducted on subadult fish paired for 0, 1, 2, 3, 4, 5, and 16 months. Our findings reveal that alterations in cyp19 gene expression coincide with a pairing period starting after 3 months. Both cyp19a1a and cyp19a1b expression levels were significantly elevated in paired females compared with their male counterparts and unpaired controls. Histological investigations demonstrated that sex conversion to females occurred during the 3-month pairing period. This study highlights the crucial role of cyp19a1a and cyp19a1b in the sex change process of A. ocellaris and indicates that a minimum of 5 months of pairing is necessary for completing the sex change.
Collapse
Affiliation(s)
- J Thamnawasolos
- Burapha University, Faculty of Science, Department of Biology, Chonburi, Thailand
| | - C Boonphakdee
- Burapha University, Faculty of Science, Department of Biology, Chonburi, Thailand
| |
Collapse
|
2
|
Repeated controlled ovarian stimulation-induced ovarian and uterine damage in mice through the PI3K/AKT signaling pathway. Hum Cell 2023; 36:234-243. [PMID: 36441500 DOI: 10.1007/s13577-022-00829-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022]
Abstract
The effects of repeated controlled ovarian stimulation (COS) on the female reproductive system are still controversial. This study investigated the effects of repeated COS on the ovaries and uterus of mice and its possible mechanism. Female ICR (Institute of Cancer Research) mice were subjected to the COS using pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) for 1, 3, 5, and 7 cycles. Serum hormone levels, reactive oxidative stress (ROS), 8-hydroxy-2'-deoxyguanosine (8-OHdG), total antioxidant capacity (T-AOC), and superoxide dismutase (SOD) in the mouse ovary and uterus were analyzed by ELISA. The morphology of the ovary and endometrium, ovarian apoptosis, and expressions of the vascular endothelial growth factor (VEGF), leukemia inhibitory factor (LIF), PI3K, AKT, Bax, and Bcl-2 in the ovarian and uterine tissues were tested by hematoxylin-eosin (HE) staining, immunohistochemistry, and western blot. The results showed that repeated COS significantly decreased the hormone level (estradiol, progesterone and anti-Müllerian hormone), high-quality of the MII oocyte ratio, oocyte and embryo number, antioxidant capacity (T-AOC, SOD activity), and the protein level of Bcl-2, LIF, and VEGF, but increased the oxidative damage (ROS, 8-OHdG content), embryo fragment ratio, and expression of pro-apoptotic protein Bax. In addition, the expressions of p-PI3K and p-AKT also decreased with the increase of COS cycle. In conclusion, repeated COS causes ovarian and uterus damage possibly through the PI3K/AKT signaling pathway, and this finding may provide some experimental basis for guiding clinical treatment.
Collapse
|
3
|
LIF and bFGF enhanced chicken primordial follicle activation by Wnt/β-catenin pathway. Theriogenology 2021; 176:1-11. [PMID: 34555602 DOI: 10.1016/j.theriogenology.2021.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
The cytokines leukemia inhibitory factor (LIF) and basic fibroblast growth factor (bFGF) are closely related to the development of primordial follicles. In this study, the functions and correlation of LIF and bFGF in the development of chicken primordial follicles were examined, along with the signaling pathways including protein kinase B (AKT), extracellular regulated protein kinase (ERK) and Wnt/β-catenin signaling pathways. Ovarian tissues were collected from four-day-old chicks and incubated with LIF and bFGF alone or in combination for three days to observe the changes in follicular development. Results showed that there was a time-dependent correlation between the changes in expression of LIF/its receptor (LIFR) and the developmental process of primordial follicles. LIF and bFGF exerted a synergistic effect on the activation of primordial follicles. However, SC144 (an antagonist of LIFR) inhibited this stimulating action. The effect by LIF and bFGF were shown to operate at AKT and ERK signaling pathways to suppress cell apoptosis and promote proliferation (P < 0.05) via the Wnt/β-catenin signaling (P < 0.05). In conclusion, local cytokines LIF and bFGF functioned to enhance the activation of chicken primordial follicles by increasing cell proliferation and decreasing apoptosis in the ovary involving AKT, ERK and Wnt/β-catenin signaling.
Collapse
|
4
|
Martínez-Chacón G, Yatkin E, Polari L, Deniz Dinç D, Peuhu E, Hartiala P, Saarinen N, Mäkelä S. CC chemokine ligand 2 (CCL2) stimulates aromatase gene expression in mammary adipose tissue. FASEB J 2021; 35:e21536. [PMID: 33913559 DOI: 10.1096/fj.201902485rrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/10/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Obesity is a risk factor for postmenopausal breast cancer. Obesity-related inflammation upregulates aromatase expression, the rate-limiting enzyme for estrogen synthesis, in breast adipose tissue (BAT), increasing estrogen production and cancer risk. The regulation of aromatase gene (CYP19A1) in BAT is complex, and the mechanisms linking obesity and aromatase dysregulation are not fully understood. An obesity-associated factor that could regulate aromatase is the CC chemokine ligand (CCL) 2, a pro-inflammatory factor that also activates signaling pathways implicated in CYP19A1 transcription. By using human primary breast adipose stromal cells (ASCs) and aromatase reporter (hARO-Luc) mouse mammary adipose explants, we demonstrated that CCL2 enhances the glucocorticoid-mediated CYP19A1 transcription. The potential mechanism involves the activation of PI.4 via ERK1/2 pathway. We also showed that CCL2 contributes to the pro-inflammatory milieu and aromatase expression in obesity, evidenced by increased expression of CCL2 and CYP19A1 in mammary tissues from obese hARO-Luc mice, and subcutaneous adipose tissue from obese women. In summary, our results indicate that postmenopausal obesity may promote CCL2 production in BAT, leading to exacerbation of the menopause-related inflammatory state and further stimulation of local aromatase and estrogens. These results provide new insights into the regulation of aromatase and may aid in finding approaches to prevent breast cancer.
Collapse
Affiliation(s)
- Gabriela Martínez-Chacón
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Emrah Yatkin
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Central Animal Laboratory, University of Turku, Turku, Finland
| | - Lauri Polari
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Defne Deniz Dinç
- Institute of Biomedicine, University of Turku, Turku, Finland.,FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Emilia Peuhu
- Institute of Biomedicine, University of Turku, Turku, Finland.,FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Pauliina Hartiala
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Plastic and General Surgery, Turku University Hospital (TYS), Turku, Finland
| | - Niina Saarinen
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Sari Mäkelä
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| |
Collapse
|
5
|
Gorsic LK, Dapas M, Legro RS, Hayes MG, Urbanek M. Functional Genetic Variation in the Anti-Müllerian Hormone Pathway in Women With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2019; 104:2855-2874. [PMID: 30786001 PMCID: PMC6543512 DOI: 10.1210/jc.2018-02178] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/15/2019] [Indexed: 01/08/2023]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is a highly heritable, common endocrine disorder characterized by hyperandrogenism, irregular menses, and polycystic ovaries. PCOS is often accompanied by elevated levels of anti-Müllerian hormone (AMH). AMH inhibits follicle maturation. AMH also inhibits steroidogenesis through transcriptional repression of CYP17A1. We recently identified 16 rare PCOS-specific pathogenic variants in AMH. OBJECTIVE To test whether additional members of the AMH signaling pathway also contribute to the etiology of PCOS. PARTICIPANTS/DESIGN Targeted resequencing of coding and regulatory regions of AMH and its specific type 2 receptor, AMHR2, was performed on 608 women affected with PCOS and 142 reproductively normal control women. Prediction tools of deleteriousness and in silico evidence of epigenetic modification were used to prioritize variants for functional evaluation. Dual-luciferase reporter assays and splicing assays were used to measure the impact of genetic variants on function. RESULTS We identified 20 additional variants in/near AMH and AMHR2 with significantly reduced signaling activity in in vitro assays. Collectively, from our previous study and as reported herein, we have identified a total of 37 variants with impaired activity in/near AMH and AMHR2 in 41 women affected with PCOS, or 6.7% of our PCOS cohort. Furthermore, no functional variants were observed in the 142 phenotyped controls. The functional variants were significantly associated with PCOS in our cohort of 608 women with PCOS and 142 controls (P = 2.3 × 10-5) and very strongly associated with PCOS relative to a larger non-Finnish European (gnomAD) population-based control cohort (P < 1 × 10-9). CONCLUSION The AMH signaling cascade plays an important role in PCOS etiology.
Collapse
Affiliation(s)
- Lidija K Gorsic
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Matthew Dapas
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Richard S Legro
- Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - M Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Anthropology, Northwestern University, Evanston, Illinois
| | - Margrit Urbanek
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Correspondence and Reprint Requests: Margrit Urbanek, PhD, Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 15-717, Chicago, Illinois 60611. E-mail:
| |
Collapse
|
6
|
Bhardwaj P, Au CC, Benito-Martin A, Ladumor H, Oshchepkova S, Moges R, Brown KA. Estrogens and breast cancer: Mechanisms involved in obesity-related development, growth and progression. J Steroid Biochem Mol Biol 2019; 189:161-170. [PMID: 30851382 PMCID: PMC6502693 DOI: 10.1016/j.jsbmb.2019.03.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 12/21/2022]
Abstract
Obesity is a risk factor for estrogen receptor-positive (ER+) breast cancer after menopause. The pro-proliferative effects of estrogens are well characterized and there is a growing body of evidence to also suggest an important role in tumorigenesis. Importantly, obesity not only increases the risk of breast cancer, but it also increases the risk of recurrence and cancer-associated death. Aromatase is the rate-limiting enzyme in estrogen biosynthesis and its expression in breast adipose stromal cells is hypothesized to drive the growth of breast tumors and confer resistance to endocrine therapy in obese postmenopausal women. The molecular regulation of aromatase has been characterized in response to many obesity-related molecules, including inflammatory mediators and adipokines. This review is aimed at providing an overview of our current knowledge in relation to the regulation of estrogens in adipose tissue and their role in driving breast tumor development, growth and progression.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, USA; Graduate School of Medical Sciences, Weill Cornell Medicine, New York, USA
| | - CheukMan C Au
- Department of Medicine, Weill Cornell Medicine, New York, USA
| | | | - Heta Ladumor
- Department of Medicine, Weill Cornell Medicine, New York, USA; Weill Cornell Medicine - Qatar, Doha, Qatar
| | | | - Ruth Moges
- Department of Medicine, Weill Cornell Medicine, New York, USA
| | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, USA; Graduate School of Medical Sciences, Weill Cornell Medicine, New York, USA; Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
7
|
|
8
|
Anderson RL, Ingman WV, Britt KL. Editorial: How Reproductive History Influences Our Breast Cancer Risk. Front Oncol 2017; 7:289. [PMID: 29376022 PMCID: PMC5770630 DOI: 10.3389/fonc.2017.00289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/13/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Robin L Anderson
- Metastasis Research Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, LaTrobe University, Bundoora, VIC, Australia
| | - Wendy V Ingman
- School of Medicine at The Queen Elizabeth Hospital, University of Adelaide, Adelaide, SA, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Kara L Britt
- Breast Cancer Risk and Prevention, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Early and late effects of aspirin and naproxen on microRNAs in the lung and blood of mice, either unexposed or exposed to cigarette smoke. Oncotarget 2017; 8:85716-85748. [PMID: 29156752 PMCID: PMC5689642 DOI: 10.18632/oncotarget.20464] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022] Open
Abstract
We recently showed that nonsteroidal anti-inflammatory drugs (NSAIDs) are able to inhibit the lung tumors induced by cigarette smoke, either mainstream (MCS) or environmental (ECS), in female mice. We used subsets of mice to analyze the expression of 1135 microRNAs in both lung and blood serum, as related to the whole-body exposure to smoke and/or oral administration of either aspirin or naproxen. In a first study, we evaluated early microRNA alterations in A/J mice exposed to ECS for 10 weeks, starting at birth, and/or treated with NSAIDs for 6 weeks, starting after weaning. At that time, when no histopathological change were apparent, ECS caused a considerable downregulation of pulmonary microRNAs affecting both adaptive mechanisms and disease-related pathways. Aspirin and naproxen modulated, with intergender differences, the expression of microRNAs having a variety of functions, also including regulation of cyclooxygenases and inflammation. In a second study, we evaluated late microRNA alterations in Swiss H mice exposed to MCS during the first 4 months of life and treated with NSAIDs after weaning until 7.5 months of life, when tumors were detected in mouse lung. Modulation of pulmonary microRNAs by the two NSAIDs was correlated with their ability to prevent preneoplastic lesions (microadenomas) and adenomas in the lung. In both studies, exposure to smoke and/or treatment with NSAIDs also modulated microRNA profiles in the blood serum. However, their levels were poorly correlated with those of pulmonary microRNAs, presumably because circulating microRNAs reflect the contributions from multiple organs and not only from lung.
Collapse
|
10
|
Au CC, Furness JB, Brown KA. Ghrelin and Breast Cancer: Emerging Roles in Obesity, Estrogen Regulation, and Cancer. Front Oncol 2017; 6:265. [PMID: 28119851 PMCID: PMC5220482 DOI: 10.3389/fonc.2016.00265] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/12/2016] [Indexed: 01/26/2023] Open
Abstract
Local and systemic factors have been shown to drive the growth of breast cancer cells in postmenopausal obese women, who have increased risk of estrogen receptor-positive breast cancer. Estrogens, produced locally in the breast fat by the enzyme aromatase, have an important role in promoting cancer cell proliferation. Ghrelin, a 28-amino acid peptide hormone, may also influence cancer growth. This peptide is produced in the stomach and acts centrally to regulate appetite and growth hormone release. Circulating levels of ghrelin, and its unacylated form, des-acyl ghrelin, are almost always inversely correlated with obesity, and these peptide hormones have recently been shown to inhibit adipose tissue aromatase expression. Ghrelin and des-acyl ghrelin have also been shown to be produced by some tumor cells and influence tumor growth. The ghrelin/des-acyl ghrelin–cancer axis is complex, one reason being that tumor cells have been shown to express splice variants of ghrelin, and ghrelin and des-acyl ghrelin might act at receptors other than the cognate ghrelin receptor, growth hormone secretagogue receptor 1a, in tumors. Effects of ghrelin and des-acyl ghrelin on energy homeostasis may also affect tumor development and growth. This review will summarize our current understanding of the role of ghrelin and des-acyl ghrelin in hormone-dependent cancers, breast cancer in particular.
Collapse
Affiliation(s)
- CheukMan Cherie Au
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne and Florey Institute of Neuroscience and Mental Health , Parkville, VIC , Australia
| | - Kristy A Brown
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
11
|
Jha T, Adhikari N, Halder AK, Saha A. Ligand- and Structure-Based Drug Design of Non-Steroidal Aromatase Inhibitors (NSAIs) in Breast Cancer. Oncology 2017. [DOI: 10.4018/978-1-5225-0549-5.ch004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aromatase is a multienzyme complex overexpressed in breast cancer and responsible for estrogen production. It is the potential target for designing anti-breast cancer drugs. Ligand and Structure-Based Drug Designing approaches (LBDD and SBDD) are involved in development of active and more specific Nonsteroidal Aromatase Inhibitors (NSAIs). Different LBDD and SBDD approaches are presented here to understand their utility in designing novel NSAIs. It is observed that molecules should possess a five or six membered heterocyclic nitrogen containing ring to coordinate with heme portion of aromatase for inhibition. Moreover, one or two hydrogen bond acceptor features, hydrophobicity, and steric factors may play crucial roles for anti-aromatase activity. Electrostatic, van der Waals, and p-p interactions are other important factors that determine binding affinity of inhibitors. HQSAR, LDA-QSAR, GQSAR, CoMFA, and CoMSIA approaches, pharmacophore mapping followed by virtual screening, docking, and dynamic simulation may be effective approaches for designing new potent anti-aromatase molecules.
Collapse
|
12
|
Editorial overview: Endocrine and metabolic diseases: Adipocyte dysfunction fuels signalings for breast cancer progression. Curr Opin Pharmacol 2016; 31:vii-x. [PMID: 27876260 DOI: 10.1016/j.coph.2016.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
13
|
Zahid H, Simpson ER, Brown KA. Inflammation, dysregulated metabolism and aromatase in obesity and breast cancer. Curr Opin Pharmacol 2016; 31:90-96. [PMID: 27875786 DOI: 10.1016/j.coph.2016.11.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 12/17/2022]
Abstract
Obesity is associated with an increased risk of estrogen-dependent breast cancer after menopause. Adipose tissue undergoes important changes in obesity due to excess storage of lipids, leading to adipocyte cell death and the recruitment of macrophages. The resultant state of chronic low-grade inflammation is associated with the activation of NFkB signaling and elevated levels of aromatase, the rate-limiting enzyme in estrogen biosynthesis. This occurs not only in the visceral and subcutaneous fat, but also in the breast fat. The regulation of aromatase in the breast adipose stromal cell in response to inflammatory mediators is under the control of complex signaling pathways, including metabolic pathways involving LKB1/AMPK, p53, HIF1α and PKM2. Interventions aimed at modifying weight, including diet and exercise, are associated with changes in adipose tissue inflammation and estrogen production that are likely to impact breast cancer risk. This review will present an overview of these topics.
Collapse
Affiliation(s)
- Heba Zahid
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia; Monash University, Clayton, Victoria, Australia
| | - Evan R Simpson
- Centre for Endocrinology and Metabolism, Hudson Institute for Medical Research, Clayton, Victoria, Australia; Monash University, Clayton, Victoria, Australia
| | - Kristy A Brown
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
14
|
De León-Nava MA, Álvarez-Delgado C, Donis-Maturano L, Hernández-Ruiz J, Manjarrez-Reyna AN, Cruz-Avilés E, Leon-Cabrera S, Morales-Montor J, Fragoso JM, Escobedo G. A non-hepatotropic parasite infection increases mortality in the acetaminophen-induced acute liver failure murine model: possible roles for IL-5 and IL-6. Mem Inst Oswaldo Cruz 2016; 111:757-764. [PMID: 27812602 PMCID: PMC5146742 DOI: 10.1590/0074-02760160311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/24/2016] [Indexed: 11/22/2022] Open
Abstract
We evaluated the effects of a non-hepatotropic parasite infection (Taenia crassiceps) on the outcome of acetaminophen-induced acute liver failure in mice. Uninfected and T. crassiceps infected mice orally received either 300 mg/kg acetaminophen or water as vehicle (n = 5 per group). Survival analysis, hepatocyte necrosis, alanine aminotransferase (ALT) levels, CYP2E1 protein, interleukin (IL-) 5, and IL-6 were assessed for all groups. All infected mice died within 16 h after exposure to acetaminophen (Tc+APAP group), whereas only one-third of uninfected animals exposed to acetaminophen (APAP group) died. Uninfected (Control group) and infected (Tc group) mice that received the vehicle showed no liver damage. Tc+APAP mice exhibited massive liver necrosis characterised by marked balloning degeneration of hepatocytes and higher serum ALT compared to Control, Tc, and APAP animals. Liver tissue from Tc+APAP mice also displayed increased expression of CYP2E1 protein and higher mRNA and protein levels of IL-5 and IL-6 compared to the other groups. These findings suggest that non-hepatotropic parasite infections may increase mortality following acute liver failure by promoting hepatocyte necrosis via IL-5 and IL-6-dependent CYP2E1 overproduction. This study identifies new potential risk factors associated with severe acute liver failure in patients.
Collapse
Affiliation(s)
- Marco A De León-Nava
- Centro de Investigación Científica y de Educación Superior de Ensenada, Departamento de Innovación Biomédica, Baja California, México
| | - Carolina Álvarez-Delgado
- Centro de Investigación Científica y de Educación Superior de Ensenada, Departamento de Innovación Biomédica, Baja California, México
| | - Luis Donis-Maturano
- Centro de Investigación Científica y de Educación Superior de Ensenada, Departamento de Innovación Biomédica, Baja California, México
| | - Joselin Hernández-Ruiz
- Universidad Nacional Autónoma de México, Facultad de Medicina, Unidad de Investigación en Medicina Experimental, Hospital General de México Dr Eduardo Liceaga, Laboratorio de Hígado, Páncreas y Motilidad, Ciudad de México, México
| | - Aaron N Manjarrez-Reyna
- Universidad Nacional Autónoma de México, Facultad de Medicina, Unidad de Investigación en Medicina Experimental, Hospital General de México Dr Eduardo Liceaga, Laboratorio de Hígado, Páncreas y Motilidad, Ciudad de México, México
| | - Edgar Cruz-Avilés
- Universidad Nacional Autónoma de México, Facultad de Medicina, Unidad de Investigación en Medicina Experimental, Hospital General de México Dr Eduardo Liceaga, Laboratorio de Hígado, Páncreas y Motilidad, Ciudad de México, México
| | - Sonia Leon-Cabrera
- Universidad Nacional Autónoma de México, Facultad de Estudios Superiores-Iztacala, Unidad de Biomedicina, Carrera de Médico Cirujano, Los Reyes Iztacala, México
| | - Jorge Morales-Montor
- Universidad Nacional Autónoma de México, Instituto de Investigaciones Biomédicas, Departamento de Inmunología, Ciudad de México, México
| | - José M Fragoso
- Instituto Nacional de Cardiología Ignacio Chávez, Departamento de Biología Molecular, Ciudad de México, México
| | - Galileo Escobedo
- Universidad Nacional Autónoma de México, Facultad de Medicina, Unidad de Investigación en Medicina Experimental, Hospital General de México Dr Eduardo Liceaga, Laboratorio de Hígado, Páncreas y Motilidad, Ciudad de México, México
| |
Collapse
|
15
|
Subbaramaiah K, Brown KA, Zahid H, Balmus G, Weiss RS, Herbert BS, Dannenberg AJ. Hsp90 and PKM2 Drive the Expression of Aromatase in Li-Fraumeni Syndrome Breast Adipose Stromal Cells. J Biol Chem 2016; 291:16011-23. [PMID: 27467582 PMCID: PMC4965552 DOI: 10.1074/jbc.m115.698902] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/23/2016] [Indexed: 11/06/2022] Open
Abstract
Li-Fraumeni syndrome (LFS) patients harbor germ line mutations in the TP53 gene and are at increased risk of hormone receptor-positive breast cancers. Recently, elevated levels of aromatase, the rate-limiting enzyme for estrogen biosynthesis, were found in the breast tissue of LFS patients. Although p53 down-regulates aromatase expression, the underlying mechanisms are incompletely understood. In the present study, we found that LFS stromal cells expressed higher levels of Hsp90 ATPase activity and aromatase compared with wild-type stromal cells. Inhibition of Hsp90 ATPase suppressed aromatase expression. Silencing Aha1 (activator of Hsp90 ATPase 1), a co-chaperone of Hsp90 required for its ATPase activity, led to both inhibition of Hsp90 ATPase activity and reduced aromatase expression. In comparison with wild-type stromal cells, increased levels of the Hsp90 client proteins, HIF-1α, and PKM2 were found in LFS stromal cells. A complex comprised of HIF-1α and PKM2 was recruited to the aromatase promoter II in LFS stromal cells. Silencing either HIF-1α or PKM2 suppressed aromatase expression in LFS stromal cells. CP-31398, a p53 rescue compound, suppressed levels of Aha1, Hsp90 ATPase activity, levels of PKM2 and HIF-1α, and aromatase expression in LFS stromal cells. Consistent with these in vitro findings, levels of Hsp90 ATPase activity, Aha1, HIF-1α, PKM2, and aromatase were increased in the mammary glands of p53 null versus wild-type mice. PKM2 and HIF-1α were shown to co-localize in the nucleus of stromal cells of LFS breast tissue. Taken together, our results show that the Aha1-Hsp90-PKM2/HIF-1α axis mediates the induction of aromatase in LFS.
Collapse
Affiliation(s)
- Kotha Subbaramaiah
- From the Department of Medicine, Weill Cornell Medical College, New York, New York 10065,
| | - Kristy A Brown
- the Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Heba Zahid
- the Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia, Monash University, Clayton, Victoria 3800, Australia, the Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia
| | - Gabriel Balmus
- the Department of Biomedical Sciences, Cornell University, Ithaca, New York 14853, and
| | - Robert S Weiss
- the Department of Biomedical Sciences, Cornell University, Ithaca, New York 14853, and
| | - Brittney-Shea Herbert
- the Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Andrew J Dannenberg
- From the Department of Medicine, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
16
|
Chan HJ, Petrossian K, Chen S. Structural and functional characterization of aromatase, estrogen receptor, and their genes in endocrine-responsive and -resistant breast cancer cells. J Steroid Biochem Mol Biol 2016; 161:73-83. [PMID: 26277097 PMCID: PMC4752924 DOI: 10.1016/j.jsbmb.2015.07.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 11/22/2022]
Abstract
Aromatase and estrogen receptor α (ER) are two key proteins for the proliferation of endocrine-responsive and -resistant breast cancers. Aromatase is an enzyme involved in the conversion of androgen (such as testosterone) to estrogen (such as 17β-estradiol). It is also a very effective therapeutic target for the treatment of endocrine-responsive breast cancer. Comparing endocrine-responsive and -resistant breast cancer, aromatase protein levels do not change significantly. Aromatase activity; however, can be increased via PI3K/Akt/IGFR signaling pathways in endocrine resistant cells. The activity of aromatase has been reported to be modulated by phosphorylation. The ER is an important steroid nuclear receptor in the proliferation of both endocrine-responsive and -resistant cells. Although the mutation or amplification of ER can cause endocrine resistance, it is not commonly found. Some point mutations and translocation events have been characterized and shown to promote estrogen-independent growth. Phosphorylation by cross-talk with growth factor pathways is one of the main mechanisms for ligand-independent activation of ER. Taken together, both ER and aromatase are important in ER-dependent breast cancer and the development of endocrine resistance.
Collapse
Affiliation(s)
- Hei Jason Chan
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States.
| |
Collapse
|
17
|
Argolo DF, Iyengar NM, Hudis CA. Obesity and Cancer: Concepts and Challenges. Indian J Surg Oncol 2015; 6:390-8. [PMID: 27081257 DOI: 10.1007/s13193-015-0483-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/17/2015] [Indexed: 12/17/2022] Open
Abstract
The rates of overweight and obesity are increasing worldwide in both developed and developing countries. Obesity is a major public health problem as it is associated with many diseases, including diabetes, hypertension, dyslipidemia, atherosclerosis, and some types of cancer. Breast cancer is a malignancy in which both the risk of development and the prognosis are negatively impacted by the obese state. The precise mechanisms pathophysiologically linking obesity and cancer are still under investigation. The biological basis for these associations includes both systemic and local tissue effects and white adipose tissue inflammation appears to be a critical component. A comprehensive understanding of the mechanisms linking obesity, inflammation and cancer may provide an opportunity for the development of strategies to attenuate the negative impact of obesity.
Collapse
Affiliation(s)
- Daniel F Argolo
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street - 8th floor, New York, NY 10065 USA ; CLION - CAM Group, Salvador, BA Brazil
| | - Neil M Iyengar
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street - 8th floor, New York, NY 10065 USA ; Weill Cornell Medical College, New York, NY USA
| | - Clifford A Hudis
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street - 8th floor, New York, NY 10065 USA ; Weill Cornell Medical College, New York, NY USA
| |
Collapse
|
18
|
Polari L, Yatkin E, Martínez Chacón MG, Ahotupa M, Smeds A, Strauss L, Zhang F, Poutanen M, Saarinen N, Mäkelä SI. Weight gain and inflammation regulate aromatase expression in male adipose tissue, as evidenced by reporter gene activity. Mol Cell Endocrinol 2015; 412:123-30. [PMID: 26054748 DOI: 10.1016/j.mce.2015.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/11/2015] [Accepted: 06/01/2015] [Indexed: 11/19/2022]
Abstract
Obesity and white adipose tissue (WAT) inflammation are associated with enhanced aromatization in women, but little is known about the regulation of aromatase (CYP19A1) gene expression in male WAT. We investigated the impact of weight gain and WAT inflammation on the regulation of CYP19A1 in males, by utilizing the hARO-Luc aromatase reporter mouse model containing a >100-kb 5'-region of the human CYP19A1 gene. We show that hARO-Luc reporter activity is enhanced in WAT of mice with increased adiposity and inflammation. Dexamethasone and TNFα, as well as forskolin and phorbol 12-myristate 13-acetate, upregulate hARO-Luc activity, suggesting the involvement of promoters I.4 and I.3/II. Furthermore, we show that diet enriched with antioxidative plant polyphenols attenuates WAT inflammation and hARO-Luc activity in obese males. In conclusion, our data suggest that obesity-associated WAT inflammation leads to increased peripheral CYP19A1 expression in males, and that polyphenol-enriched diet may have the potential to attenuate excessive aromatization in WAT of obese men.
Collapse
Affiliation(s)
- L Polari
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - E Yatkin
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - M G Martínez Chacón
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - M Ahotupa
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - A Smeds
- Åbo Akademi University, Process Chemistry Centre, Laboratory of Wood and Paper Chemistry, Turku, Finland
| | - L Strauss
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - F Zhang
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - M Poutanen
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - N Saarinen
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - S I Mäkelä
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
19
|
Jha T, Adhikari N, Halder AK, Saha A. Ligand- and Structure-Based Drug Design of Non-Steroidal Aromatase Inhibitors (NSAIs) in Breast Cancer. QUANTITATIVE STRUCTURE-ACTIVITY RELATIONSHIPS IN DRUG DESIGN, PREDICTIVE TOXICOLOGY, AND RISK ASSESSMENT 2015. [DOI: 10.4018/978-1-4666-8136-1.ch011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Aromatase is a multienzyme complex overexpressed in breast cancer and responsible for estrogen production. It is the potential target for designing anti-breast cancer drugs. Ligand and Structure-Based Drug Designing approaches (LBDD and SBDD) are involved in development of active and more specific Nonsteroidal Aromatase Inhibitors (NSAIs). Different LBDD and SBDD approaches are presented here to understand their utility in designing novel NSAIs. It is observed that molecules should possess a five or six membered heterocyclic nitrogen containing ring to coordinate with heme portion of aromatase for inhibition. Moreover, one or two hydrogen bond acceptor features, hydrophobicity, and steric factors may play crucial roles for anti-aromatase activity. Electrostatic, van der Waals, and p-p interactions are other important factors that determine binding affinity of inhibitors. HQSAR, LDA-QSAR, GQSAR, CoMFA, and CoMSIA approaches, pharmacophore mapping followed by virtual screening, docking, and dynamic simulation may be effective approaches for designing new potent anti-aromatase molecules.
Collapse
|
20
|
To SQ, Knower KC, Cheung V, Simpson ER, Clyne CD. Transcriptional control of local estrogen formation by aromatase in the breast. J Steroid Biochem Mol Biol 2015; 145:179-86. [PMID: 24846828 DOI: 10.1016/j.jsbmb.2014.05.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/11/2014] [Indexed: 12/11/2022]
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'.
Collapse
Affiliation(s)
- Sarah Q To
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia.
| | - Kevin C Knower
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia.
| | - Vanessa Cheung
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia
| | - Evan R Simpson
- Metabolism and Cancer Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia
| | - Colin D Clyne
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
21
|
To SQ, Cheung V, Lazarus KA, Knower KC, Clyne CD. Estradiol regulates Tumor Necrosis Factor-α expression and secretion in Estrogen Receptor positive breast cancer cells. Mol Cell Endocrinol 2014; 394:21-8. [PMID: 25004254 DOI: 10.1016/j.mce.2014.06.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 06/17/2014] [Accepted: 06/30/2014] [Indexed: 12/20/2022]
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-β-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.
Collapse
Affiliation(s)
- Sarah Q To
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia
| | - Vanessa Cheung
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia
| | - Kyren A Lazarus
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia; Environmental and Biotechnology Centre, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
| | - Kevin C Knower
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia.
| | - Colin D Clyne
- Cancer Drug Discovery Laboratory, MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
22
|
Abstract
Prostate cancer is the commonest, non-cutaneous cancer in men. At present, there is no cure for the advanced, castration-resistant form of the disease. Estrogen has been shown to be important in prostate carcinogenesis, with evidence resulting from epidemiological, cancer cell line, human tissue and animal studies. The prostate expresses both estrogen receptor alpha (ERA) and estrogen receptor beta (ERB). Most evidence suggests that ERA mediates the harmful effects of estrogen in the prostate, whereas ERB is tumour suppressive, but trials of ERB-selective agents have not translated into improved clinical outcomes. The role of ERB in the prostate remains unclear and there is increasing evidence that isoforms of ERB may be oncogenic. Detailed study of ERB and ERB isoforms in the prostate is required to establish their cell-specific roles, in order to determine if therapies can be directed towards ERB-dependent pathways. In this review, we summarise evidence on the role of ERB in prostate cancer and highlight areas for future research.
Collapse
Affiliation(s)
- Adam W Nelson
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - Wayne D Tilley
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - David E Neal
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - Jason S Carroll
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| |
Collapse
|
23
|
Kinoshita T, Honma S, Shibata Y, Yamashita K, Watanabe Y, Maekubo H, Okuyama M, Takashima A, Takeshita N. An innovative LC-MS/MS-based method for determining CYP 17 and CYP 19 activity in the adipose tissue of pre- and postmenopausal and ovariectomized women using 13C-labeled steroid substrates. J Clin Endocrinol Metab 2014; 99:1339-47. [PMID: 24456285 DOI: 10.1210/jc.2013-3715] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Does adipose tissue produce steroid hormones like an endocrine organ? OBJECT To clarify whether adipose tissue produces sex steroid hormone like an endocrine organ, we estimated several key steroid hormone levels, as well as CYP17 and CYP19 activity, in ovariectomized, pre- and postmenopausal women by liquid chromatography-tandem mass spectrometry (LC-MS/MS). SUBJECTS AND METHODS The subjects were 19 premenopausal (n = 12), postmenopausal (n = 4), and ovariectomized women (n = 3) aged 27-68 years. Serum, visceral adipose and sc adipose samples were taken from these subjects and stored at -70°C. The levels of cortisol, cortisone, progesterone (Prog), androstenedione, dehydroepiandrosterone, estrone, estradiol (E2), and T in serum and adipose tissue were estimated simultaneously by LC-MS/MS. CYP17 and CYP19 activity in tissues were assayed with the use of (13)C-labeled steroid precursors and LC-MS/MS-based estimation of the metabolites. RESULTS E2 and Prog levels in the sera of postmenopausal or ovariectomized women were less than 10% of those in premenopausal women. No marked variations were seen in other hormones. Estrone, androstenedione, dehydroepiandrosterone, and Prog levels in the visceral and sc tissues of postmenopausal and ovariectomized women were 9-60 times higher than those in serum, whereas E2 and T levels were 3- to 7-fold higher than those in serum, and cortisol and cortisone levels were 20% of those found for serum. CYP17 in adipose tissue was found to have 17-hydroxylase and 20,17-lyase activity, with each catalytic activity being essentially equal. Therefore, CYP17 in adipose tissue is of the testicular/ovarian type but not adrenal type, which has 17-hydroxylase activity dominant. The presence of CYP19 activity in adipose tissue was approximately 3% of CYP17. CONCLUSION Our findings suggest that adipose tissue acts as an endocrine organ, with CYP17 and CYP19 activity playing an essential role in sex steroid hormone biosynthesis.
Collapse
Affiliation(s)
- Toshihiko Kinoshita
- Department of Obstetrics and Gynecology (T.K., A.T., N.T.), Toho University Medical Center Sakura Hospital, Chiba 285-8741, Japan; Department of Analytical Research (S.H., Y.W., H.M., M.O.), ASKA Pharma Medical Co Ltd, Kawasaki 213-8522, Japan; Department of Urology (Y.S.), Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; and Department of Clinical Analysis (K.Y.), Tohoku Pharmaceutical University, Sendai 981-8558, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Knutson TP, Lange CA. Tracking progesterone receptor-mediated actions in breast cancer. Pharmacol Ther 2014; 142:114-25. [PMID: 24291072 PMCID: PMC3943696 DOI: 10.1016/j.pharmthera.2013.11.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 11/15/2013] [Indexed: 12/13/2022]
Abstract
Ovarian steroid hormones contribute to breast cancer initiation and progression primarily through the actions of their nuclear transcription factors, the estrogen receptor alpha (ERα) and progesterone receptors (PRs). These receptors are important drivers of the luminal A and B subtypes of breast cancer, where estrogen-blocking drugs have been effective endocrine therapies for patients with these tumors. However, many patients do not respond, or become resistant to treatment. When endocrine therapies fail, the luminal subtypes of breast cancer are more difficult to treat because these subtypes are among the most heterogeneous in terms of mutation diversity and gene expression profiles. Recent evidence suggests that progestin and PR actions may be important drivers of luminal breast cancers. Clinical trial data has demonstrated that hormone replacement therapy with progestins drives invasive breast cancer and results in greater mortality. PR transcriptional activity is dependent upon cross-talk with growth factor signaling pathways that alter PR phosphorylation, acetylation, or SUMOylation as mechanisms for regulating PR target gene selection required for increased cell proliferation and survival. Site-specific PR phosphorylation is the primary driver of gene-selective PR transcriptional activity. However, PR phosphorylation and heightened transcriptional activity is coupled to rapid PR protein degradation; the range of active PR detected in tumors is likely to be dynamic. Thus, PR target gene signatures may provide a more accurate means of tracking PR's contribution to tumor progression rather than standard clinical protein-based (IHC) assays. Further development of antiprogestin therapies should be considered alongside antiestrogens and aromatase inhibitors.
Collapse
Affiliation(s)
- Todd P Knutson
- Departments of Medicine, Division of Hematology, Oncology, and Transplantation and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carol A Lange
- Departments of Medicine, Division of Hematology, Oncology, and Transplantation and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
25
|
In subfertile couple, abdominal fat loss in men is associated with improvement of sperm quality and pregnancy: a case-series. PLoS One 2014; 9:e86300. [PMID: 24520319 PMCID: PMC3919721 DOI: 10.1371/journal.pone.0086300] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 12/08/2013] [Indexed: 12/13/2022] Open
Abstract
Background The impact of overweight among men of reproductive-age may affect fertility. Abdominal fat, more than body mass index, is an indicator of higher metabolic risk, which seems to be involved in decreasing sperm quality. This study aims to assess the relationship between abdominal fat and sperm DNA fragmentation and the effect of abdominal fat loss, among 6 men in subfertile couples. Methods Sperm DNA fragmentation, abdominal fat and metabolic and hormonal profiles were measured in the 6 men before and after dietary advices. Seminal oxidative stress and antioxidant markers were determined. Results After several months of a lifestyle program, all 6 men lost abdominal fat (patient 1: loss of 3 points of abdominal fat, patient 2: loss of 3 points, patient 3: loss of 2 points, patient 4: loss of 1 point, patient 5: loss of 4 points and patient 6: loss of 13 points). At the same time, their rate of sperm DNA fragmentation decreased: 9.5% vs 31%, 24% vs 43%, 18% vs 47%, 26.3% vs 66%, 25.4% vs 35% and 1.7% vs 25%. Also, an improvement in both metabolic (significant decrease in triglycerides and total cholesterol; p = 0.0139) and hormonal (significant increase in testosterone/oestradiol ratio; p = 0.0139) blood profiles was observed after following the lifestyle program. In seminal plasma, the amount of SOD2 has significantly increased (p = 0.0139) while in parallel carbonylated proteins have decreased. Furthermore, all spouses got pregnant. All pregnancies were brought to term. Conclusion This study shows specifically that sperm DNA fragmentation among men in subfertile couples could be affected by abdominal fat, but improvement of lifestyle factor may correct this alteration. The effect of specific abdominal fat loss on sperm quality needs further investigation. The reduction of oxidative stress may be a contributing factor.
Collapse
|
26
|
Yamaguchi Y, Seino Y, Takei H, Kurosumi M, Hayashi SI. Detection of estrogen-independent growth-stimulating activity in breast cancer tissues: implication for tumor aggressiveness. CANCER MICROENVIRONMENT 2013; 7:23-31. [PMID: 24203105 PMCID: PMC4150880 DOI: 10.1007/s12307-013-0139-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/24/2013] [Indexed: 01/02/2023]
Abstract
Estrogen and various growth factors affecting tumor behavior are present in the breast cancer microenvironment, but their comprehensive effects and signal crosstalks are different in each case. However, there is no system to evaluate the factors, detected in individual breast cancer cases, that regulate ER activity and tumor progression. In this study, we analyzed the effects of individual breast cancer extracts by our original system using an estrogen-signal reporter cell line, MCF-7-E10, which we previously established. MCF-7-E10 cell line is stably transfected by an estrogen response element (ERE)-green fluorescent protein (GFP) gene; it expresses GFP when estrogen receptors (ERs) are activated by estrogen or growth factor signal-mediated ER phosphorylation. Using this cell line, we analyzed the comprehensive effects of factors derived from breast cancer tissues on ER activity and growth of MCF-7-E10 cells for each case. We also analyzed relationships between these activities and clinicopathologic characteristics of patients who provided cancer specimens. The breast cancer extracts, which reflect the combined activities of growth factors present in individual cases, stimulated MCF-7-E10 cell growth in an estrogen-independent manner, and specifically stimulated growth of other breast cancer cell lines, regardless of ER expression. High growth-promoting activities were seen in tumor regions of specimens with tumors > 10 mm in size, HER2 intrinsic subtype, and scirrhous and solid-tubular carcinoma histological subtypes. Anti-human hepatocyte growth factor (HGF) antibody and an inhibitor for insulin-like growth factor-1 (IGF-1) receptor inhibited MCF-7-E10 cell growth by the breast cancer extracts, indicating that signal pathways via HGF or IGF-1 receptor significantly affect breast cancer. These data suggest that growth factors other than estrogen in the tumor extract significantly affect breast cancer aggressiveness in an estrogen-independent manner, and could be useful therapeutic targets.
Collapse
Affiliation(s)
- Yuri Yamaguchi
- Research Institute for Clinical Oncology, Saitama Cancer Center, 818 Komuro, Ina-machi, Saitama-ken, 362-0806, Japan,
| | | | | | | | | |
Collapse
|
27
|
Gautier A, Bonnet F, Dubois S, Massart C, Grosheny C, Bachelot A, Aubé C, Balkau B, Ducluzeau PH. Associations between visceral adipose tissue, inflammation and sex steroid concentrations in men. Clin Endocrinol (Oxf) 2013; 78:373-8. [PMID: 22469460 DOI: 10.1111/j.1365-2265.2012.04401.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/18/2012] [Accepted: 03/30/2012] [Indexed: 11/28/2022]
Abstract
CONTEXT In men, obesity and the metabolic syndrome are accompanied by decreased testosterone levels, but little is known about the associations between visceral adipose tissue (VAT), VAT-related inflammation and sex steroids. OBJECTIVE To examine the relative impact of VAT, abdominal subcutaneous adipose tissue (SAT) and interleukin 6 (IL-6), a marker of VAT-induced inflammation, on testosterone (T) and 17β-oestradiol (E2) levels in dysmetabolic men. METHODS We study the NUMEVOX cohort of 229 men, aged 27-77 years, who all had at least one metabolic syndrome criterion (on average three). IL-6, C-reactive protein, Homeostasis Model Assessment of (HOMA) insulin resistance index (HOMA-IR), liver enzymes, E2, LH, sex hormone-binding globulin (SHBG), T, waist circumference and body mass index (BMI) were measured; bioavailable testosterone (BT) was calculated from T and SHBG; MRI-assessed VAT and SAT were analysed in 109 of these men. RESULTS Visceral adipose tissue was strongly correlated with E2 (Spearman r = 0.38, P < 0.001) and with BT/E2 ratio (r = -0.42, P < 0.001), while SAT was not correlated with either. IL-6 was correlated with E2 (r = 0.19, P = 0.007), BT (r = -0.19, P = 0.006) and BT/E2 ratio (r = -0.30 P < 0.001). In multivariate linear analysis, the relation between VAT and E2 was independent of age, BMI (P = 0.008), leptin (P < 0.001), T and SHBG. Log(IL-6) was significantly inversely related with log(BT) (P = 0.032) independently of age, VAT, leptin and HOMA-IR. CONCLUSIONS 17β-oestradiol levels were positively associated with VAT, but not with SAT, while T and BT were negatively and independently associated with IL-6. The significant inverse association between IL-6 and T suggests an important role of low-grade visceral fat inflammation in the central hypogonadism associated with the metabolic syndrome.
Collapse
Affiliation(s)
- Alain Gautier
- Département d'Endocrinologie-Diabétologie-Nutrition, CHU Rennes, Université Rennes1, Hôpital Sud, Rennes, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
To SQ, Knower KC, Clyne CD. NFκB and MAPK signalling pathways mediate TNFα-induced Early Growth Response gene transcription leading to aromatase expression. Biochem Biophys Res Commun 2013; 433:96-101. [DOI: 10.1016/j.bbrc.2013.02.058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 02/10/2013] [Indexed: 12/23/2022]
|
29
|
Involvement of early growth response factors in TNFα-induced aromatase expression in breast adipose. Breast Cancer Res Treat 2013; 138:193-203. [PMID: 23338760 DOI: 10.1007/s10549-013-2413-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 01/10/2013] [Indexed: 12/20/2022]
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.
Collapse
|
30
|
To SQ, Takagi K, Miki Y, Suzuki K, Abe E, Yang Y, Sasano H, Simpson ER, Knower KC, Clyne CD. Epigenetic mechanisms regulate the prostaglandin E receptor 2 in breast cancer. J Steroid Biochem Mol Biol 2012; 132:331-8. [PMID: 22929011 DOI: 10.1016/j.jsbmb.2012.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/18/2012] [Accepted: 07/31/2012] [Indexed: 01/16/2023]
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.
Collapse
MESH Headings
- Adipose Tissue/cytology
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Line, Tumor
- CpG Islands
- DNA Methylation
- Decitabine
- Epigenesis, Genetic
- 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/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Reference Values
- Stromal Cells/metabolism
Collapse
Affiliation(s)
- Sarah Q To
- Cancer Drug Discovery Laboratory, Prince Henry's Institute of Medical Research, Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Quignot N, Arnaud M, Robidel F, Lecomte A, Tournier M, Cren-Olivé C, Barouki R, Lemazurier E. Characterization of endocrine-disrupting chemicals based on hormonal balance disruption in male and female adult rats. Reprod Toxicol 2012; 33:339-52. [DOI: 10.1016/j.reprotox.2012.01.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 01/11/2012] [Accepted: 01/12/2012] [Indexed: 10/14/2022]
|
32
|
A comparison of two human cell lines and two rat gonadal cell primary cultures as in vitro screening tools for aromatase modulation. Toxicol In Vitro 2012; 26:107-18. [DOI: 10.1016/j.tiv.2011.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 10/25/2011] [Accepted: 11/09/2011] [Indexed: 11/20/2022]
|
33
|
Armani A, Marzolla V, Rosano GMC, Fabbri A, Caprio M. Phosphodiesterase type 5 (PDE5) in the adipocyte: a novel player in fat metabolism? Trends Endocrinol Metab 2011; 22:404-11. [PMID: 21741267 DOI: 10.1016/j.tem.2011.05.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 05/18/2011] [Accepted: 05/25/2011] [Indexed: 12/19/2022]
Abstract
Phosphodiesterase type 5 (PDE5) is expressed in many tissues (e.g. heart, lung, pancreas, penis) and plays a specific role in hydrolyzing cyclic guanosine monophosphate (cGMP). In adipocytes, cGMP regulates crucial functions by activating cGMP-dependent protein kinase (PKG). Interestingly, PDE5 was recently identified in adipose tissue, although its role remains unclear. Its inhibition, however, was recently shown to affect adipose differentiation and aromatase function. This review summarizes evidence supporting a role for the PDE5-regulated cGMP/PKG system in adipose tissue and its effects on adipocyte function. A better elucidation of the role of PDE5 in the adipocyte could reveal new therapeutic strategies for fighting obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Andrea Armani
- Center for Clinical and Basic Research, Scientific Institute for Research, Hospitalization and Health Care (IRCCS) San Raffaele Pisana, Rome, Italy
| | | | | | | | | |
Collapse
|
34
|
Khan SI, Zhao J, Khan IA, Walker LA, Dasmahapatra AK. Potential utility of natural products as regulators of breast cancer-associated aromatase promoters. Reprod Biol Endocrinol 2011; 9:91. [PMID: 21693041 PMCID: PMC3142499 DOI: 10.1186/1477-7827-9-91] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 06/21/2011] [Indexed: 12/21/2022] Open
Abstract
Aromatase, the key enzyme in estrogen biosynthesis, converts androstenedione to estrone and testosterone to estradiol. The enzyme is expressed in various tissues such as ovary, placenta, bone, brain, skin, and adipose tissue. Aromatase enzyme is encoded by a single gene CYP 19A1 and its expression is controlled by tissue-specific promoters. Aromatase mRNA is primarily transcribed from promoter I.4 in normal breast tissue and physiological levels of aromatase are found in breast adipose stromal fibroblasts. Under the conditions of breast cancer, as a result of the activation of a distinct set of aromatase promoters (I.3, II, and I.7) aromatase expression is enhanced leading to local overproduction of estrogen that promotes breast cancer. Aromatase is considered as a potential target for endocrine treatment of breast cancer but due to nonspecific reduction of aromatase activity in other tissues, aromatase inhibitors (AIs) are associated with undesirable side effects such as bone loss, and abnormal lipid metabolism. Inhibition of aromatase expression by inactivating breast tumor-specific aromatase promoters can selectively block estrogen production at the tumor site. Although several synthetic chemical compounds and nuclear receptor ligands are known to inhibit the activity of the tumor-specific aromatase promoters, further development of more specific and efficacious drugs without adverse effects is still warranted. Plants are rich in chemopreventive agents that have a great potential to be used in chemotherapy for hormone dependent breast cancer which could serve as a source for natural AIs. In this brief review, we summarize the studies on phytochemicals such as biochanin A, genistein, quercetin, isoliquiritigenin, resveratrol, and grape seed extracts related to their effect on the activation of breast cancer-associated aromatase promoters and discuss their aromatase inhibitory potential to be used as safer chemotherapeutic agents for specific hormone-dependent breast cancer.
Collapse
Affiliation(s)
- Shabana I Khan
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
- Department of Pharmacognosy, University of Mississippi, University, MS 38677, USA
| | - Jianping Zhao
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Ikhlas A Khan
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
- Department of Pharmacognosy, University of Mississippi, University, MS 38677, USA
| | - Larry A Walker
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
- Department of Pharmacology, University of Mississippi, MS 38677, USA
- University of Mississippi Cancer Institute, University of Mississippi, University, MS 38677, USA
| | - Asok K Dasmahapatra
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
- Department of Pharmacology, University of Mississippi, MS 38677, USA
| |
Collapse
|
35
|
Chen D, Reierstad S, Fang F, Bulun SE. JunD and JunB integrate prostaglandin E2 activation of breast cancer-associated proximal aromatase promoters. Mol Endocrinol 2011; 25:767-75. [PMID: 21393445 DOI: 10.1210/me.2010-0368] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Aromatase is the key enzyme in estrogen biosynthesis. Normal breast adipose tissue expresses low levels of aromatase via the distal promoter I.4. Breast adipose tissue surrounding a tumor exhibits excessive aromatase expression controlled by proximal aromatase promoters I.3/II, leading to high local levels of estrogen and breast cancer progression. Prostaglandin E(2) (PGE(2)) secreted by malignant breast epithelial cells activates breast cancer-associated aromatase promoters I.3/II, but silences promoter I.4, in cultured human breast adipose fibroblasts (BAF). The c-Jun N-terminal kinase 1 and p38α mitogen activated protein kinases are necessary for PGE(2) activation of aromatase promoters I.3/II; thus, we examined the roles of downstream targets, c-Jun, JunB, JunD, and activating transcription factor 2, in PGE(2)-mediated regulation of aromatase expression in BAF. PGE(2) induced JunB and JunD protein expression through protein kinase A and protein kinase C, respectively. JunB or JunD knockdown by small interfering RNA markedly reduced PGE(2)-induced total aromatase mRNA level and enzyme activity via promoters I.3/II. JunB knockdown also abrogated JunD expression. JunB stimulated, whereas JunD inhibited, aromatase promoter I.4 activity. Activating transcription factor 2 knockdown did not affect promoter-specific or total aromatase mRNA levels. c-Jun knockdown increased promoter I.4-specific and PGE(2)-induced promoters I.3/II-specific aromatase mRNA levels, leading to enhanced PGE(2)-induced total aromatase mRNA level and enzyme activity. JunD, c-Jun, and JunB bound to a CRE(-211/-199) essential for PGE(2) induction of aromatase promoters I.3/II. Taken together, JunD and c-Jun repress aromatase promoter I.4. JunD mediates, whereas c-Jun modulates, PGE(2) activation of aromatase promoters I.3/II via CRE(-211/-199). JunB also activates aromatase promoters I.3/II by maintaining JunD expression. Targeting JunD may abolish aromatase expression selectively in breast cancer tissue.
Collapse
Affiliation(s)
- Dong Chen
- Division of Reproductive Biology Research, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA.
| | | | | | | |
Collapse
|
36
|
Subbaramaiah K, Howe LR, Bhardwaj P, Du B, Gravaghi C, Yantiss RK, Zhou XK, Blaho VA, Hla T, Yang P, Kopelovich L, Hudis CA, Dannenberg AJ. Obesity is associated with inflammation and elevated aromatase expression in the mouse mammary gland. Cancer Prev Res (Phila) 2011; 4:329-46. [PMID: 21372033 PMCID: PMC3071249 DOI: 10.1158/1940-6207.capr-10-0381] [Citation(s) in RCA: 302] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elevated circulating estrogen levels are associated with increased risk of breast cancer in obese postmenopausal women. Following menopause, the biosynthesis of estrogens through CYP19 (aromatase)-mediated metabolism of androgen precursors occurs primarily in adipose tissue, and the resulting estrogens are then secreted into the systemic circulation. The potential links between obesity, inflammation, and aromatase expression are unknown. In both dietary and genetic models of obesity, we observed necrotic adipocytes surrounded by macrophages forming crown-like structures (CLS) in the mammary glands and visceral fat. The presence of CLS was associated with activation of NF-κB and increased levels of proinflammatory mediators (TNF-α, IL-1β, Cox-2), which were paralleled by elevated levels of aromatase expression and activity in the mammary gland and visceral fat of obese mice. Analyses of the stromal-vascular and adipocyte fractions of the mammary gland suggested that macrophage-derived proinflammatory mediators induced aromatase and estrogen-dependent gene expression (PR, pS2) in adipocytes. Saturated fatty acids, which have been linked to obesity-related inflammation, stimulated NF-κB activity in macrophages leading to increased levels of TNF-α, IL-1β, and Cox-2, each of which contributed to the induction of aromatase in preadipocytes. The discovery of the obesity → inflammation → aromatase axis in the mammary gland and visceral fat and its association with CLS may provide insight into mechanisms underlying the increased risk of hormone receptor-positive breast cancer in obese postmenopausal women, the reduced efficacy of aromatase inhibitors in the treatment of breast cancer in these women, and their generally worse outcomes. The presence of CLS may be a biomarker of increased breast cancer risk or poor prognosis.
Collapse
Affiliation(s)
- Kotha Subbaramaiah
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Louise R. Howe
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Baoheng Du
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Claudia Gravaghi
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Rhonda K. Yantiss
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Xi Kathy Zhou
- Department of Public Health, Weill Cornell Medical College, New York, New York
| | - Victoria A. Blaho
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Timothy Hla
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Peiying Yang
- Department of General Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Levy Kopelovich
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Clifford A. Hudis
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | |
Collapse
|
37
|
Subbaramaiah K, Hudis CA, Dannenberg AJ. The prostaglandin transporter regulates adipogenesis and aromatase transcription. Cancer Prev Res (Phila) 2011; 4:194-206. [PMID: 21212407 DOI: 10.1158/1940-6207.capr-10-0367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cytochrome P450 aromatase, encoded by the CYP19 gene, catalyzes estrogen synthesis. In obese postmenopausal women, increased estrogen synthesis in adipose tissue has been linked to hormone-dependent breast carcinogenesis. Hence, it is important to elucidate the mechanisms that regulate CYP19 gene expression. Prostaglandin E(2) (PGE(2)) stimulates the cyclic AMP (cAMP) → protein kinase A (PKA) → cAMP responsive element binding protein (CREB) pathway leading to increased CYP19 transcription. The prostaglandin transporter (PGT) removes PGE(2) from the extracellular milieu and delivers it to the cytosol, where it is inactivated. The main objective of this study was to determine whether PGT regulates CYP19 transcription. Silencing of PGT in preadipocytes increased PGE(2) levels in the extracellular medium, thereby stimulating the cAMP → PKA pathway resulting in enhanced interaction between pCREB, p300, and the CYP19 I.3/II promoter. A reciprocal decrease in the interaction between the CYP19 I.3/II promoter and BRCA1, a repressor of CYP19 transcription, was observed. Overexpressing PGT reduced extracellular PGE(2) levels, suppressed the cAMP → PKA pathway, enhanced the interaction between BRCA1 and p300, and inhibited aromatase expression. We also compared the PGT → aromatase axis in preadipocytes versus adipocytes. Aromatase levels were markedly increased in preadipocytes versus adipocytes. This increase in aromatase was explained, at least in part, by reduced PGT levels leading to enhanced PGE(2) → cAMP → PKA signaling. In addition to regulating aromatase expression, PGT-mediated changes in extracellular PGE(2) levels were a determinant of adipocyte differentiation. Collectively, these results suggest that PGT modulates adipogenesis and thereby PGE(2)-mediated activation of the cAMP → PKA → CREB pathway leading to altered CYP19 transcription and aromatase activity.
Collapse
Affiliation(s)
- Kotha Subbaramaiah
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW To review the recent literature on the involvement and importance of leukemia inhibitory factor (LIF) in the human implantation process, and the attempts using LIF-based interventions to improve assisted reproductive technologies (ARTs) outcome in women with recurrent implantation failure. RECENT FINDINGS High LIF expression is an indicator of receptive endometrium in fertile women. However, in infertile individuals, the data on endometrial LIF expression and secretion are controversial. Even after ruling out other causes of infertility, such as tubal, endocrine, male factor, and endometriosis, LIF-only detection is not sufficient for assessment of implantation potential in women with unexplained infertility. This is obviously in contrast to evidence of the crucial role of LIF in mouse endometrial physiology. In a large multicenter study, recombinant human LIF failed to improve the outcome of IVF treatment in women with recurrent implantation failure. SUMMARY A better comprehension of the mechanisms underlying endometrial receptivity and implantation should guide clinicians through proper management and treatment of infertility and implantation failure, and may eventually enable widespread adherence to single embryo transfer practices.
Collapse
|
39
|
Brueggemeier RW, Su B, Darby MV, Sugimoto Y. Selective regulation of aromatase expression for drug discovery. J Steroid Biochem Mol Biol 2010; 118:207-10. [PMID: 19931613 PMCID: PMC3638894 DOI: 10.1016/j.jsbmb.2009.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 11/05/2009] [Accepted: 11/15/2009] [Indexed: 10/20/2022]
Abstract
Aromatase is a particularly attractive drug target in the treatment of hormone-responsive breast cancer, and aromatase activity in breast cancer patients is greater in or near the tumor tissue compared with the normal breast tissue. Complex regulation of aromatase expression in human tissues involves alternative promoter sites that provide tissue-specific control. Previous studies in our laboratories suggested a strong association between aromatase (CYP19) gene expression and the expression of cyclooxygenase (COX) genes. Additionally, COX selective inhibitors can suppress CYP19 gene expression and decrease aromatase activity. Our current hypothesis is that pharmacological regulation of aromatase can act locally to decrease the biosynthesis of estrogen and may provide additional therapy options for patients with hormone-dependent breast cancer. Two pharmacological approaches are being developed, one approach utilizing small molecule drug design and the second approach involving mRNA silencing technology. The small molecule drug design approach focuses on the synthesis and biological evaluation of a novel series of sulfonanilide analogs derived from COX-2 selective inhibitors. Combinatorial chemistry approaches were used to generate diversely substituted novel sulfonanilides. The compounds suppress aromatase enzyme activity in SK-BR-3 breast cancer cells in a dose and time dependent manner, and structure activity analysis does not find a correlation between aromatase suppression and COX inhibition. Real-time PCR analysis demonstrates that the sulfonanilide analogs decrease aromatase gene transcription in breast cells. Furthermore, the sulfonanilide compounds selectively decrease aromatase gene expression in several breast cancer cells, without exhibiting cytotoxic or apoptotic effects at low micromole concentrations. A ligand-based pharmacophore model for selective aromatase modulation (SAM) by the novel sulfonanilides identified an aromatic ring, two hydrogen bond acceptors, and a hydrophobic function as four key chemical features. In the second approach, short interfering RNAs (siRNA) were designed targeting human aromatase mRNA. Treatment of breast cancer cells with siRNAs targeting aromatase (siAROMs) completely masked the aromatase enzyme activity and resulted in suppression of CYP19 mRNA. Thus, these results suggest that the novel sulfonanilides and the siRNAs targeting aromatase expression may be valuable tools for selective regulation of aromatase in breast cancer.
Collapse
|
40
|
Kim JY, Han EH, Kim HG, Oh KN, Kim SK, Lee KY, Jeong HG. Bisphenol A-induced aromatase activation is mediated by cyclooxygenase-2 up-regulation in rat testicular Leydig cells. Toxicol Lett 2010; 193:200-8. [PMID: 20096755 DOI: 10.1016/j.toxlet.2010.01.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 01/11/2010] [Accepted: 01/13/2010] [Indexed: 02/04/2023]
Abstract
Bisphenol A (4,4'-dihydroxy-2,2-diphenylpropane; BPA) is an endocrine disruptor that affects the reproductive health of wildlife and possibly of humans. Evidence suggests that BPA interrupts ovarian steroidogenesis by altering steroidogenic enzymes. We evaluated the effect of BPA on aromatase expression in rat testicular Leydig cells. In addition, we investigated whether cyclooxygenase-2 (COX-2) was involved in BPA-induced aromatase expression. BPA induced a time- and concentration-dependent increase in aromatase protein expression in rat testicular Leydig R2C cells. It also increased aromatase gene expression and its enzyme and promoter activity, but reduced testosterone synthesis; increased COX-2 mRNA expression and promoter activity, the production of prostaglandin E(2) (PGE(2)), and the gene expression of PGE(2) (EP2 and EP4) receptors; induced the activation of cyclic adenosine monophosphate (cAMP) response element (CRE) and CREB binding; and increased the phosphorylation of protein kinase A (PKA), Akt, and mitogen-activated protein (MAP) kinase signaling pathways. BPA activation of aromatase was reversed by various inhibitors (COX-2, PKA, Akt, ERK, JNK, and p38). Taken together, these results suggest that BPA increases aromatase activity, which is correlated with COX-2 up-regulation mediated by the CRE, PKA, Akt, and MAP kinase signaling pathways in rat testicular Leydig cells.
Collapse
Affiliation(s)
- Ji Young Kim
- College of Pharmacy, Research Institute of Drug Development, Chonnam National University, Gwangju, South Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Saldanha CJ, Duncan KA, Walters BJ. Neuroprotective actions of brain aromatase. Front Neuroendocrinol 2009; 30:106-18. [PMID: 19450619 PMCID: PMC2700852 DOI: 10.1016/j.yfrne.2009.04.016] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 04/02/2009] [Accepted: 04/14/2009] [Indexed: 12/16/2022]
Abstract
The steroidal regulation of vertebrate neuroanatomy and neurophysiology includes a seemingly unending list of brain areas, cellular structures and behaviors modulated by these hormones. Estrogens, in particular have emerged as potent neuromodulators, exerting a range of effects including neuroprotection and perhaps neural repair. In songbirds and mammals, the brain itself appears to be the site of injury-induced estrogen synthesis via the rapid transcription and translation of aromatase (estrogen synthase) in astroglia. This induction seems to occur regardless of the nature and location of primary brain damage. The induced expression of aromatase apparently elevates local estrogen levels enough to interfere with apoptotic pathways, thereby decreasing secondary degeneration and ultimately lessening the extent of damage. There is even evidence suggesting that aromatization may affect injury-induced cytogenesis. Thus, aromatization in the brain appears to confer neuroprotection by an array of mechanisms that involve the deceleration and acceleration of degeneration and repair, respectively. We are only beginning to understand the factors responsible for the injury-induced transcription of aromatase in astroglia. In contrast, much of the manner in which local and circulating estrogens may achieve their neuroprotective effects has been elucidated. However, gaps in our knowledge include issues about the cell-specific regulation of aromatase expression, steroidal influences of aromatization distinct from estrogen formation, and questions about the role of constitutive aromatase in neuroprotection. Here we describe the considerable consensus and some interesting differences in knowledge gained from studies conducted on diverse animal models, experimental paradigms and preparations towards understanding the neuroprotective actions of brain aromatase.
Collapse
Affiliation(s)
- Colin J Saldanha
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015, United States.
| | | | | |
Collapse
|
42
|
Li YF, Hu W, Fu SQ, Li JD, Liu JH, Kavanagh JJ. Aromatase inhibitors in ovarian cancer: is there a role? Int J Gynecol Cancer 2008; 18:600-14. [PMID: 17894799 DOI: 10.1111/j.1525-1438.2007.01075.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Estrogen plays a role in ovarian tumorigenesis. Aromatase is the enzyme required for the synthesis of estrogen via conversion of androgen to estrogen, which is the major source of estrogen in postmenopausal women. Aromatase is present in normal ovaries and other tissues (e.g., fat and muscle) as well as in 33-81% tumor tissues of ovarian cancer. Aromatase inhibitors (AIs) block estrogen synthesis by inhibiting aromatase activity. In patients with recurrent ovarian cancer, single-agent AI therapy has been shown to elicit clinical response rates of up to 35.7% and stable disease rates of 20-42%. Given the limited treatment options for recurrent ovarian cancer and the favorable safety profile and convenient use, AI is a rational option for prolonging platinum-free interval in recurrent ovarian cancer. Further studies are required to determine the efficacy of combination treatment with AIs and biological agents, determine the benefit of AIs for treating special types of ovarian cancer (e.g., endometrioid type), and identify biomarkers for targeted patient selection. This review summarizes the current epidemiologic, preclinical, and clinical data regarding estrogen's role in ovarian cancer, the expression and regulation of aromatase in this disease, the development and characteristics of the three generations of AIs, and the preclinical and clinical studies of AIs in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Y F Li
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | | | | | | | | | | |
Collapse
|
43
|
Bukulmez O, Hardy DB, Carr BR, Word RA, Mendelson CR. Inflammatory status influences aromatase and steroid receptor expression in endometriosis. Endocrinology 2008; 149:1190-204. [PMID: 18048499 PMCID: PMC2275353 DOI: 10.1210/en.2007-0665] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aberrant up-regulation of aromatase in eutopic endometrium and implants from women with endometriosis has been reported. Aromatase induction may be mediated by increased cyclooxygenase-2 (COX-2). Recently, we demonstrated that progesterone receptor (PR)-A and PR-B serve an antiinflammatory role in the uterus by antagonizing nuclear factor kappaB activation and COX-2 expression. PR-C, which antagonizes PR-B, is up-regulated by inflammation. Although estrogen receptor alpha (ERalpha) is implicated in endometriosis, an antiinflammatory role of ERbeta has been suggested. We examined stage-specific expression of aromatase, COX-2, ER, and PR isoform expression in eutopic endometrium, implants, peritoneum, and endometrioma samples from endometriosis patients. Endometrial and peritoneal biopsies were obtained from unaffected women and those with fibroids. Aromatase expression in eutopic endometrium from endometriosis patients was significantly increased compared with controls. Aromatase expression in endometriosis implants was markedly increased compared with eutopic endometrium. Aromatase mRNA levels were increased significantly in red implants relative to black implants and endometrioma cyst capsule. Moreover, COX-2 expression was increased in implants and in eutopic endometrium of women with endometriosis as compared with control endometrium. As observed for aromatase mRNA, the highest levels of COX-2 mRNA were found in red implants. The ratio of ERbeta/ERalpha mRNA was significantly elevated in endometriomas compared with endometriosis implants and eutopic endometrium. Expression of PR-C mRNA relative to PR-A and PR-B mRNA was significantly increased in endometriomas compared with eutopic and control endometrium. PR-A protein was barely detectable in endometriomas. Thus, whereas PR-C may enhance disease progression, up-regulation of ERbeta may play an antiinflammatory and opposing role.
Collapse
Affiliation(s)
- Orhan Bukulmez
- Department of Obstetrics, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9032, USA
| | | | | | | | | |
Collapse
|
44
|
Subbaramaiah K, Hudis C, Chang SH, Hla T, Dannenberg AJ. EP2 and EP4 receptors regulate aromatase expression in human adipocytes and breast cancer cells. Evidence of a BRCA1 and p300 exchange. J Biol Chem 2008; 283:3433-3444. [PMID: 18083712 DOI: 10.1074/jbc.m705409200] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023] Open
Abstract
Cytochrome P450 aromatase (aromatase), a product of the CYP19 gene, catalyzes the synthesis of estrogens from androgens. Because aromatase-dependent estrogen biosynthesis has been linked to hormone-dependent breast carcinogenesis, it is important to elucidate the mechanisms that regulate CYP19 gene expression. The main objective of this study was to identify the receptors (EP) for prostaglandin E(2) (PGE(2)) that mediate the induction of CYP19 transcription in human adipocytes and breast cancer cells. Treatment with PGE(2) induced aromatase, an effect that was mimicked by either EP(2) or EP(4) agonists. Antagonists of EP(2) or EP(4) or small interference RNA-mediated down-regulation of these receptors suppressed PGE(2)-mediated induction of aromatase. PGE(2) via EP(2) and EP(4) stimulated the cAMP-->protein kinase A pathway resulting in enhanced interaction between P-CREB, p300, and the aromatase promoter I.3/II. Overexpressing a mutant form of p300 that lacks histone acetyltransferase activity suppressed PGE(2)-mediated induction of aromatase promoter activity. PGE(2) via EP(2) and EP(4) also caused a reduction in both the amounts of BRCA1 and the interaction between BRCA1 and the aromatase promoter I.3/II. Activation of the aromatase promoter by PGE(2) was suppressed by overexpressing wild-type BRCA1. Silencing of EP(2) or EP(4) also blocked PGE(2)-mediated induction of the progesterone receptor, a prototypic estrogen-response gene. In a mouse model, overexpressing COX-2 in the mammary gland, a known inducer of PGE(2) synthesis, led to increased aromatase mRNA and activity and reduced amounts of BRCA1; these effects were reversed by knocking out EP(2). Taken together, these results suggest that PGE(2) via EP(2) and EP(4) activates the cAMP-->PKA-->CREB pathway leading to enhanced CYP19 transcription and increased aromatase activity. Reciprocal changes in the interaction between BRCA1, p300, and the aromatase promoter I.3/II contributed to the inductive effects of PGE(2).
Collapse
MESH Headings
- Adipocytes/enzymology
- Adipocytes/metabolism
- Animals
- Aromatase/biosynthesis
- Aromatase/genetics
- BRCA1 Protein/genetics
- Breast Neoplasms/enzymology
- Cell Line, Tumor
- Dinoprostone/metabolism
- E1A-Associated p300 Protein/genetics
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Humans
- Mammary Neoplasms, Animal/metabolism
- Mice
- Mice, Transgenic
- Receptors, Prostaglandin E/metabolism
- Receptors, Prostaglandin E, EP2 Subtype
- Receptors, Prostaglandin E, EP4 Subtype
Collapse
Affiliation(s)
- Kotha Subbaramaiah
- Department of Medicine, Weill Cornell Medical College, New York, New York 10065.
| | - Clifford Hudis
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Sung-Hee Chang
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut 06030-3501
| | - Timothy Hla
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut 06030-3501
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
45
|
McInnes KJ, Brown KA, Knower KC, Chand AL, Clyne CD, Simpson ER. Characterisation of aromatase expression in the human adipocyte cell line SGBS. Breast Cancer Res Treat 2008; 112:429-35. [DOI: 10.1007/s10549-007-9883-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 12/20/2007] [Indexed: 10/22/2022]
|
46
|
Su B, Díaz-Cruz ES, Landini S, Brueggemeier RW. Suppression of aromatase in human breast cells by a cyclooxygenase-2 inhibitor and its analog involves multiple mechanisms independent of cyclooxygenase-2 inhibition. Steroids 2008; 73:104-11. [PMID: 18045633 PMCID: PMC3638915 DOI: 10.1016/j.steroids.2007.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Accepted: 09/14/2007] [Indexed: 11/29/2022]
Abstract
Previous studies have demonstrated that cyclooxygenase-2 (COX-2) inhibitor NS-398 decrease aromatase activity at the transcript level in breast cancer cells. However, N-Methyl NS-398, which does not have COX-2 inhibitory activity but has very similar structure to NS-398, decreases aromatase activity and transcription in MCF-7 and MDA-MB-231 breast cells to the same extent as NS-398. This suggests that NS-398 decrease aromatase expression in breast cancer cells via other mechanism(s). Further investigations find that both compounds only decrease aromatase activity stimulated by forskolin/phorbol ester at the transcript level in both breast cancer cell lines and in breast stromal cells from patients. They do not affect aromatase expression and activity stimulated by dexamethasone. Both compounds also suppress MCF-7 cell proliferation stimulated by testosterone. Aromatase inhibition studies using placental microsomes demonstrate that the compounds show only weak direct aromatase inhibition. These results suggest that NS-398 and its N-methyl analog suppress aromatase expression and activity with multiple mechanisms.
Collapse
Affiliation(s)
| | | | | | - Robert W. Brueggemeier
- TO WHOM CORRESPONDENCE SHOULD BE ADDRESSED: Robert W. Brueggemeier. Address: 500 W. 12th Avenue, Columbus, OH, USA 43210, Telephone: 614-292-5231, Facsimile: 614-292-3113, Email address:
| |
Collapse
|
47
|
Thorat MA, Morimiya A, Mehrotra S, Konger R, Badve SS. Prostanoid receptor EP1 expression in breast cancer. Mod Pathol 2008; 21:15-21. [PMID: 17906615 DOI: 10.1038/modpathol.3800970] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cyclooxygenase enzymes play an important role in carcinogenesis, and increased expression of cyclooxygenase enzymes has been reported in cancers arising at a number of different sites. Most, if not all of these actions are thought to be mediated by prostaglandin E2 (PGE2). The actions of PGE2 are mediated via four main prostanoid receptors, designated EP1, EP2, EP3 and EP4, based on their different pharmacological properties and secondary messenger pathways. Recently, expression of EP1 has been reported in rat mammary gland and the inhibition of this receptor has been documented to have chemopreventive effect in this animal model. EP1 has also been shown to decrease the incidence of colon cancer in mouse models. In this study, we analysed the expression of EP1 in normal and malignant breast tissues. Expression of EP1 was analysed in breast (benign and cancer) cell lines by reverse-transcriptase polymerase chain reaction and by western blot analyses. Expression was also analysed by immunohistochemistry in normal breast tissues and in 89 cases of breast cancer. Semiquantitative analysis of the staining was performed. The data were compared with and correlated with other prognostic factors like tumour size, tumour grade, lymph node status, oestrogen receptor, progesterone receptor (PR), HER2/neu and cyclooxygenase-2. EP1 expression was demonstrated in human breast cancer by immunohistochemistry. Expression of EP1 was seen both in the cytoplasm and/or in the nuclear membrane in majority of cases. Nuclear EP1 expression correlated with PR (P=0.032) and inversely with node positivity (P=0.025). However, EP1 expression did not correlate with expression of cyclooxygenase-2 (P=0.059). Expression of EP1 is frequently seen in human breast cancers. Nuclear expression of EP1 correlates with good prognosis markers like node negative status and PR expression.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Blotting, Western
- Breast Neoplasms/chemistry
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cell Nucleus/chemistry
- Cyclooxygenase 2/analysis
- Cytoplasm/chemistry
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Middle Aged
- Neoplasm Staging
- Prognosis
- RNA, Messenger/analysis
- Receptor, ErbB-2/analysis
- Receptors, Estrogen/analysis
- Receptors, Progesterone/analysis
- Receptors, Prostaglandin E/analysis
- Receptors, Prostaglandin E/genetics
- Receptors, Prostaglandin E, EP1 Subtype
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Mangesh A Thorat
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
48
|
Licznerska BE, Wegman PP, Nordenskjöld B, Wingren S. In situ levels of oestrogen producing enzymes and its prognostic significance in postmenopausal breast cancer patients. Breast Cancer Res Treat 2007; 112:15-23. [PMID: 18030614 DOI: 10.1007/s10549-007-9819-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Accepted: 11/08/2007] [Indexed: 11/24/2022]
Abstract
BACKGROUND The risk of developing breast cancer is strongly correlated with the overall exposure to oestrogen and most tumours are more or less dependent on oestrogen for their growth. A great majority of breast cancers occur after menopause when the ovaries have ceased to be functional, yet breast tumours in postmenopausal women maintain high intratumoural oestrogen concentrations, primarily through enzymatic conversion of androgenic precursors. PATIENTS with a hormone dependent tumour generally receive the anti-oestrogen tamoxifen that mediate its anti-tumour effect by competing with oestrogen for binding to the oestrogen-receptor (ER). We therefore propose that the levels of oestrogen producing enzymes may affect the prognosis in postmenopausal breast cancer patients treated with tamoxifen. METHODS We measured the mRNA and protein levels of aromatase and sulfatase by real-time PCR (n=161) and immunohistochemistry (n=131) in postmenopausal women with breast cancer. RESULTS A significant better recurrence-free survival was detected in patients with weak or high protein expression of stromal aromatase (P=0.0008), as also demonstrated by a decreased relative risk (RR=0.50, CI=0.33-0.76, P=0.003). When we combined patients with weak and high stromal aromatase and selected only ER-positive patients, the improved prognosis was even more evident (P=0.0000) and was shown to be a significant prognostic factor in a multivariate Cox-model (HR=0.15, CI=0.06-0.39, P=0.000). The mRNA expression of aromatase and sulfatase, as well as the protein expression of sulfatase revealed no prognostic significance. CONCLUSION Protein expression of stromal aromatase may serve as a significant prognostic marker in ER-positive patients.
Collapse
Affiliation(s)
- Barbara E Licznerska
- Institution of Clinical and Experimental Medicine, Division of Cell Biology, Faculty of Health Sciences, Linköping University, Linkoping, Sweden
| | | | | | | |
Collapse
|
49
|
Salhab M, Singh-Ranger G, Mokbel R, Jouhra F, Jiang WG, Mokbel K. Cyclooxygenase-2 mRNA expression correlates with aromatase expression in human breast cancer. J Surg Oncol 2007; 96:424-8. [PMID: 17657731 DOI: 10.1002/jso.20740] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
INTRODUCTION The cyclooxygenase-2 (COX-2), responsible for the conversion of arachidonic acid into prostaglandin (PG) E2, is known to increase intracellular cAMP and estrogen production in malignant breast tissue. The aromatase enzyme complex is responsible for local production of estrogens in breast cancer. Increasing evidence supports a role for COX-2 in upregulation of aromatase activity. The aim of this study was to examine the relationship between COX-2 and aromatase mRNA expression in human breast cancer. METHODS A total of 160 breast samples (127 tumor tissues and 33 normal tissues) were analyzed. Levels of transcription were determined using real-time quantitative PCR. COX-2 and aromatase mRNA expression were normalized against CK19. Levels of expression of COX-2 were correlated with those of aromatase using Pearson's correlation method. RESULTS Levels of expression of COX-2/CK19 of both benign and malignant tissues were positively correlated with aromatase/CK19 transcript levels (correlation coefficient = +0.536, P < 0.0001). When we compared levels of expression of both genes in malignant samples only, there was a highly significant positive correlation (r = +0.611, P < 0.00001). CONCLUSION This study demonstrates a strong positive relationship between COX-2 and aromatase mRNA expression, and lends further support to the hypothesis that COX-2 is an upregulator of aromatase in breast tissue.
Collapse
Affiliation(s)
- M Salhab
- St. George's University of London, Tooting, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
50
|
Oestrogen producing enzymes and mammary carcinogenesis: a review. Breast Cancer Res Treat 2007; 111:191-202. [DOI: 10.1007/s10549-007-9788-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 10/05/2007] [Indexed: 10/22/2022]
|