32
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Dysregulated metabolism and the regulation of aromatase in breast adipose stromal cells in obesity and cancer

      abstract
      1 , 2 , 1 , 1 , 3 , 3 , 1 , 4 , 1 , 2 ,
      BMC Proceedings
      BioMed Central
      Metabolism, diet and disease
      29-31 May 2012

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background The risk of breast cancer in postmenopausal women is increased two-fold with obesity, and the majority of breast tumours that arise are oestrogen-dependent. After menopause, when the ovaries cease to produce oestrogens, it is the local production of oestrogens within the breast adipose stromal cells (ASCs) which promotes and sustains tumour growth. This is largely due to the increased expression of aromatase, responsible for the conversion of androgens to oestrogens. Aromatase expression in breast cancer is known to be under the control of a proximal promoter, promoter PII, which is maximally activated by cAMP-dependent mechanisms. We have previously demonstrated that the LKB1/AMPK pathway is a key negative regulator of aromatase expression within the breast by inhibiting the nuclear translocation of the CREB co-activator CRTC2 [1]. We have also demonstrated that the tumour-derived factor prostaglandin E2 (PGE2) and the obesity-associated factor leptin stimulate aromatase expression by inhibiting LKB1 and AMPK expression and activity. Hypoxia inducible factor-1α (HIF1α) is emerging as a potent regulator of glycolysis in tumour cells and we have identified a putative hypoxia response element in aromatase promoter Pll immediately adjacent the cAMP response element, known to be bound by the CREB-CRTC2 complex in ASCs in breast cancer. We therefore hypothesise that HIF1α may be involved in regulating aromatase in breast cancer. Materials and methods Primary human breast ASCs were isolated from tissue after breast reduction surgery. Real-time PCR, Western blotting, immunofluorescence and high content screening were used to assess HIF1α expression and localisation after PGE2 treatment. Chromatin immunoprecipitation (ChIP) was performed to examine the interaction of HIF1α with aromatase promoter PII and reporter assays were performed to assess the effect of HIF1α on PII activity. Double immunohistochemistry for HIF1α and aromatase was also performed on sections of formalin-fixed paraffin-embedded breast tissue from breast cancer patients and cancer-free patients. Results We have found that PGE2 increases HIF1α transcript expression, nuclear localisation and binding to aromatase PII in primary human breast ASCs. Moreover, HIF1α causes a significant increase in PII activity and acts cooperatively with CREB to cause this induction. Data from breast cancer patient samples demonstrates that HIF1α is also increased in tumour-associated ASCs compared to breast tissue from cancer-free women. Interestingly, the majority of ASCs from breast cancer patient samples display staining for both HIF1α and aromatase. Conclusions This study is part of a growing body of evidence indicating that dysregulated metabolism is not only a characteristic of adipocytes in obesity and epithelial cells in cancer, but also occurs in tumour-associated ASCs. We demonstrate that dysregulation of metabolic pathways is accompanied by an increase in aromatase expression within the breast adipose and provide an additional mechanism whereby obesity is linked to breast cancer. Clinical studies are currently underway to explore the use of drugs which target these pathways, such as metformin, for their use as novel aromatase inhibitors for the prevention and treatment of postmenopausal breast cancer.

          Related collections

          Most cited references1

          • Record: found
          • Abstract: found
          • Article: not found

          Subcellular localization of cyclic AMP-responsive element binding protein-regulated transcription coactivator 2 provides a link between obesity and breast cancer in postmenopausal women.

          Epidemiologic evidence supports a correlation between obesity and breast cancer in women. AMP-activated protein kinase plays an important role in energy homeostasis and inhibits the actions of cyclic AMP-responsive element binding protein-regulated transcription coactivator 2 (CRTC2). In postmenopausal women, the cyclic AMP-responsive element binding protein-dependent regulation of aromatase is a determinant of breast tumor formation through local production of estrogens. The present work aimed to examine the effect of adipokines on aromatase expression and identify additional mechanisms by which prostaglandin E(2) causes increased aromatase expression in human breast adipose stromal cells. Treatment of human adipose stromal cells with forskolin and phorbol 12-myristate 13-acetate (PMA), to mimic prostaglandin E(2), resulted in nuclear translocation of CRTC2. Aromatase promoter II (PII) activity assays showed that CRTC2 in addition to forskolin/PMA treatment significantly increased PII-induced activity. CRTC2 binding to PII was examined by chromatin immunoprecipitation, and forskolin/PMA treatment was associated with increased binding to PII. Treatment of human adipose stromal cells with leptin significantly up-regulated aromatase expression associated with nuclear translocation of CRTC2 and increased binding of CRTC2 to PII. Adiponectin treatment significantly decreased forskolin/PMA-stimulated aromatase expression, consistent with the decreased nuclear translocation of CRTC2 and the decreased binding of CRTC2 to PII. The expression and activity of the AMP-activated protein kinase LKB1 was examined and found to be significantly decreased following either forskolin/PMA or leptin treatment. In contrast, adiponectin significantly increased LKB1 expression and activity. In conclusion, the regulation of aromatase by CRTC2, in response to the altered hormonal milieu associated with menopause and obesity, provides a critical link between obesity and breast cancer.
            Bookmark

            Author and article information

            Conference
            BMC Proc
            BMC Proc
            BMC Proceedings
            BioMed Central
            1753-6561
            2012
            1 June 2012
            : 6
            : Suppl 3
            : P9
            Affiliations
            [1 ]Metabolism & Cancer Laboratory, Prince Henry’s Institute, Clayton, Victoria, 3168, Australia
            [2 ]Department of Physiology, Monash University, Clayton, Victoria, 3168, Australia
            [3 ]Depatment of Pathology, Tohoku University, Sendai, Japan
            [4 ]Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3168, Australia
            Article
            1753-6561-6-S3-P9
            10.1186/1753-6561-6-S3-P9
            3394444
            b4e9de15-13a8-4ec3-bb19-073b6945798e
            Copyright ©2012 Samarajeewa et al; licensee BioMed Central Ltd.

            This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

            Metabolism, diet and disease
            Washington, DC, USA
            29-31 May 2012
            History
            Categories
            Poster Presentation

            Medicine
            Medicine

            Comments

            Comment on this article