1
|
Parija M, Prakash S, Krishna BM, Dash S, Mishra SK. SIRT1 mediates breast cancer development and tumorigenesis controlled by estrogen-related receptor β. Breast Cancer 2024; 31:440-455. [PMID: 38421553 DOI: 10.1007/s12282-024-01555-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 02/11/2024] [Indexed: 03/02/2024]
Abstract
Silent mating type information regulation 2 homolog 1 (SIRT1) is a class III histone deacetylase (HDAC) that is NAD + dependent and essential for metabolism, senescence, and cell survival. SIRT1 is overexpressed in several cancers, including breast cancer. SIRT1 is a well-known target gene of the estrogen receptor alpha (ER alpha) and is closely related to ER alpha deacetylation. Transcription factor Estrogen-related receptors (ERRs) share sequence homology with ERs in the DNA-binding domain, therefore, the possibility of sharing target genes between them is high. Our current research aims to gain insight into the function of ERRβ in regulating the activity of SIRT1 during the progression of breast cancer. ER-positive (ER + ve) breast cancer cells and tissues had considerably enhanced SIRT1 expression. Six potential ERRE sites were identified by analysis of the 5' upstream region of SIRT1, and both in vitro and in vivo experiments supported their presence. We found SIRT1 to be up-regulated in ERRβ overexpressed ER + ve breast cancer cells. Furthermore, our findings suggested that ectopic production of ERR and PCAF would increase SIRT1 activity. Our findings also indicated that ectopic production of ERRβ and PCAF increased SIRT1 activity. With sufficient evidence demonstrating the substantial involvement of SIRT1 in cell proliferation, migration, and colony formation capability, we were also able to illustrate the tumorigenic role of SIRT1. Overall, our findings highlight SIRT1's tumorigenic influence on breast cancer and suggest that SIRT1 inhibitors might serve as potential therapeutic drugs for the treatment of breast cancer.
Collapse
Affiliation(s)
- Monalisa Parija
- Cancer Biology Lab, Gene Function and Regulation Group, Institute of Life Sciences, Nalco Square, Nadankanan Road, Kalinga Hospital Cross, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana (NCR Delhi), 121001, India
| | - Surya Prakash
- Cancer Biology Lab, Gene Function and Regulation Group, Institute of Life Sciences, Nalco Square, Nadankanan Road, Kalinga Hospital Cross, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana (NCR Delhi), 121001, India
| | - B Madhu Krishna
- Department of Medical Oncology, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA, 91010, USA
| | - Sanghamitra Dash
- Cancer Biology Lab, Gene Function and Regulation Group, Institute of Life Sciences, Nalco Square, Nadankanan Road, Kalinga Hospital Cross, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana (NCR Delhi), 121001, India
| | - Sandip K Mishra
- Cancer Biology Lab, Gene Function and Regulation Group, Institute of Life Sciences, Nalco Square, Nadankanan Road, Kalinga Hospital Cross, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
2
|
Spadazzi C, Mercatali L, Esposito M, Wei Y, Liverani C, De Vita A, Miserocchi G, Carretta E, Zanoni M, Cocchi C, Bongiovanni A, Recine F, Kang Y, Ibrahim T. Trefoil factor-1 upregulation in estrogen-receptor positive breast cancer correlates with an increased risk of bone metastasis. Bone 2021; 144:115775. [PMID: 33249323 DOI: 10.1016/j.bone.2020.115775] [Citation(s) in RCA: 7] [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: 05/21/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Bone is one of the most preferred sites of metastatic spread from different cancer types, including breast cancer. However, different breast cancer subtypes exhibit distinct metastatic behavior in terms of kinetics and anatomic sites of relapse. Despite advances in the diagnosis, the identification of patients at high-risk of bone recurrence is still an unmet clinical need. We conducted a retrospective analysis, by gene expression and immunohistochemical assays, on 90 surgically resected breast cancer samples collected from patients who experienced no evidence of distant metastasis, bone or visceral metastasis in order to identify a primary tumor-derived marker of bone recurrence. We identified trefoil factor-1 (pS2 or TFF1) as strictly correlated to bone metastasis from ER+ breast cancer. In silico analysis was carried out to confirm this observation, linking gene expression data with clinical characteristics available from public clinical datasets. Then, we investigated TFF1 function in ER+ breast cancer tumorigenesis and bone metastasis through xenograft in vivo models of MCF 7 breast cancer with gain and loss of function of TFF1. As a response to microenvironmental features in primary tumors, TFF1 expression could modulate ER+ breast cancer growth, leading to a less proliferative phenotype. Our results showed it may not play a role in late stages of bone metastasis, however further studies are warranted to understand whether it could contribute in the early-stages of the metastatic cascade. In conclusion, TFF1 upregulation in primary ER+ breast cancer could be useful to identify patients at high-risk of bone metastasis. This could help clinicians in the identification of patients who likely can develop bone metastasis and who could benefit from personalized treatments and follow-up strategies to prevent metastatic disease.
Collapse
Affiliation(s)
- Chiara Spadazzi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Laura Mercatali
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Chiara Liverani
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Alessandro De Vita
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Giacomo Miserocchi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | | | - Michele Zanoni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Claudia Cocchi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Federica Recine
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| |
Collapse
|
3
|
Aimo A, Vergaro G, Castiglione V, Barison A, Pasanisi E, Petersen C, Chubuchny V, Giannoni A, Poletti R, Maffei S, Januzzi JL, Passino C, Emdin M. Effect of Sex on Reverse Remodeling in Chronic Systolic Heart Failure. JACC-HEART FAILURE 2018; 5:735-742. [PMID: 28958348 DOI: 10.1016/j.jchf.2017.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/13/2017] [Accepted: 07/27/2017] [Indexed: 01/15/2023]
Abstract
OBJECTIVES This study sought to investigate sex-related differences in reverse remodeling (RR). BACKGROUND RR, that is, the recovery from left ventricular (LV) dilation and dysfunction in response to treatment for heart failure (HF), is associated with improved prognosis. METHODS Data from patients with stable systolic HF (LV ejection fraction [LVEF] of <50%) undergoing 2 transthoracic echocardiograms within 12 ± 2 months were analyzed. Reverse remodeling was defined as a ≥15% reduction in LV end-systolic volume index. RESULTS A total of 927 patients were evaluated (68 ± 12 years; median LVEF = 35% [interquartile range: 30% to 43%]; 27% women). Ischemic HF was less often encountered in women (33% vs. 60%, respectively; p < 0.001), whereas most characteristics did not differ with regard to sex. Women showed a higher incidence of RR (41% vs. 27%, respectively; p < 0.001), despite similar baseline LV volume and function. RR was more frequent among women in the subgroups with either ischemic or nonischemic HF, as well as in all categories of systolic dysfunction (LVEF ≤35% or >35%, according to current indication for device implantation, and LVEF <40% or 40% to 50% according to the definition of HF with reduced or mid-range EF). In the whole population, female sex was an independent predictor of RR (hazard ratio: 1.54; 95% confidence interval: 1.11 to 2.14; p = 0.011), together with cause of HF, disease duration, and left bundle branch block. Female sex was again an independent predictor of RR in all LVEF categories. CONCLUSIONS Reverse remodeling is more frequent among women, regardless of cause and severity of LV dysfunction. Female sex is an independent predictor of RR in all categories of LV systolic dysfunction.
Collapse
Affiliation(s)
- Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Giuseppe Vergaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | - Andrea Barison
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Emilio Pasanisi
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Christina Petersen
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | - Alberto Giannoni
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Roberta Poletti
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Silvia Maffei
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Clinical Research Institute, Boston, Massachusetts
| | - Claudio Passino
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| |
Collapse
|
4
|
Sun LN, Zhi Z, Chen LY, Zhou Q, Li XM, Gan WJ, Chen S, Yang M, Liu Y, Shen T, Xu Y, Li JM. SIRT1 suppresses colorectal cancer metastasis by transcriptional repression of miR-15b-5p. Cancer Lett 2017; 409:104-115. [PMID: 28923398 DOI: 10.1016/j.canlet.2017.09.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/29/2017] [Accepted: 09/10/2017] [Indexed: 12/19/2022]
Abstract
The class III deacetylase sirtuin 1 (SIRT1), a member of the sirtuin family proteins, plays a key role in many types of cancers including colorectal cancer (CRC). Here we report that SIRT1 suppressed CRC metastasis in vitro and in vivo as a negative regulator for miR-15b-5p transcription. Mechanistically, SIRT1 impaired regulatory effects of activator protein (AP-1) on miR-15b-5p trans-activation through deacetylation of AP-1. Importantly, acyl-CoA oxidase 1 (ACOX1), a key enzyme of the fatty acid oxidation (FAO) pathway, was found as a direct target for miR-15b-5p. SIRT1 expression was positively correlated with ACOX1 expression in CRC cells and in xenografts. Moreover, ACOX1 overexpression attenuated the augmentation of migration and invasion of CRC cells by miR-15b-5p overexpression. In conclusion, our study demonstrated a functional role of the SIRT1/miR-15b-5p/ACOX1 axis in CRC metastasis and suggested a potential target for metastatic CRC therapy.
Collapse
Affiliation(s)
- Li-Na Sun
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Zheng Zhi
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Liang-Yan Chen
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Qun Zhou
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiu-Ming Li
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Wen-Juan Gan
- First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Shu Chen
- Department of Clinical Medicine, Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Meng Yang
- Department of Clinical Medicine, Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Yao Liu
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Tong Shen
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Yong Xu
- Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, People's Republic of China.
| | - Jian-Ming Li
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
5
|
BASP1 interacts with oestrogen receptor α and modifies the tamoxifen response. Cell Death Dis 2017; 8:e2771. [PMID: 28492543 PMCID: PMC5520704 DOI: 10.1038/cddis.2017.179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 01/01/2023]
Abstract
Tamoxifen binds to oestrogen receptor α (ERα) to elicit distinct responses that vary by cell/tissue type and status, but the factors that determine these differential effects are unknown. Here we report that the transcriptional corepressor BASP1 interacts with ERα and in breast cancer cells, this interaction is enhanced by tamoxifen. We find that BASP1 acts as a major selectivity factor in the transcriptional response of breast cancer cells to tamoxifen. In all, 40% of the genes that are regulated by tamoxifen in breast cancer cells are BASP1 dependent, including several genes that are associated with tamoxifen resistance. BASP1 elicits tumour-suppressor activity in breast cancer cells and enhances the antitumourigenic effects of tamoxifen treatment. Moreover, BASP1 is expressed in breast cancer tissue and is associated with increased patient survival. Our data have identified BASP1 as an ERα cofactor that has a central role in the transcriptional and antitumourigenic effects of tamoxifen.
Collapse
|
6
|
Nakareangrit W, Thiantanawat A, Visitnonthachai D, Watcharasit P, Satayavivad J. Sodium arsenite inhibited genomic estrogen signaling but induced pERα (Ser118) via MAPK pathway in breast cancer cells. ENVIRONMENTAL TOXICOLOGY 2016; 31:1133-1146. [PMID: 25728338 DOI: 10.1002/tox.22122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 01/12/2015] [Accepted: 01/24/2015] [Indexed: 06/04/2023]
Abstract
Arsenic (As) is considered a major environmental health threat worldwide due to its widespread contamination in drinking water. Recent studies reported that arsenic is a potential xenoestrogen as it interfered with the action of estrogen (E2) and estrogen receptor (ER) signaling. The present study investigated the effects of sodium arsenite (NaAsO2 ) on estrogen signaling in human breast cancer cells. The results demonstrated that NaAsO2 dose-dependently increased viability of hormone-dependent breast cancer MCF-7 and T47D cells expressing both ERα and ERβ but not hormone-independent MDA-MB-231 cells expressing ERβ. These suggested ERα contribution to NaAsO2 -stimulated breast cancer cells growth. NaAsO2 induced down-regulation of ERα but up-regulation of ERβ protein expressions in T47D cells. Moreover, NaAsO2 dose-dependently inhibited E2-induced ER transcriptional activity as it decreased E2-mediated ERE-luciferase transcription activation and PgR mRNA transcription but increased pS2 mRNA transcription. However, NaAsO2 induced both rapid and sustained activation of ERK1/2 and increased in phosphorylation of ERα at serine 118 residue, c-fos and c-myc protein expressions. These results indicated that NaAsO2 interferes the genomic estrogen-signaling pathway but induces activation of a rapid nongenomic signal transduction through ERK1/2 pathway which may contribute to its proliferative effect on hormone-dependent breast cancer cells. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1133-1146, 2016.
Collapse
Affiliation(s)
- Watanyoo Nakareangrit
- Laboratory of Pharmacology, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
- Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
| | - Apinya Thiantanawat
- Laboratory of Pharmacology, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
- Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology, CHE, Ministry of Education, Thailand
| | - Daranee Visitnonthachai
- Laboratory of Pharmacology, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
| | - Piyajit Watcharasit
- Laboratory of Pharmacology, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
- Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology, CHE, Ministry of Education, Thailand
| | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
- Chulabhorn Graduate Institute, 54 Kamphaeng Phet 6, Talat Bang Khen, Laksi, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology, CHE, Ministry of Education, Thailand
| |
Collapse
|
7
|
Dobson LE, Fairbairn TA, Plein S, Greenwood JP. Sex Differences in Aortic Stenosis and Outcome Following Surgical and Transcatheter Aortic Valve Replacement. J Womens Health (Larchmt) 2015; 24:986-95. [PMID: 26653869 DOI: 10.1089/jwh.2014.5158] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aortic stenosis is the commonest valve defect in the developed world and is associated with a high mortality once symptomatic. There is a difference in the way that male and female hearts remodel in the face of chronic pressure overload: women develop a concentrically hypertrophied, small cavity left ventricle (LV), whereas men are more prone to the development of eccentric hypertrophy. At a cellular level, there is an increase in collagen and metalloproteinase gene expression in males suggesting a different regulation of extracellular volume composition according to sex. Male hearts with aortic stenosis appear to have more fibrosis than their female comparators. The trigger for this appears to be in part related to estrogen receptor signaling, but other factors such as renin-angiotensin activation, nitric oxide, and circulating noradrenaline levels may also be implicated. Treatment options include surgical valve replacement (SAVR) and more recently transcatheter aortic valve replacement (TAVR). Female sex may be a risk factor for adverse outcome following SAVR and conversely appears to confer a survival advantage when undergoing TAVR. Whether the lower mortality seen following TAVR in women compared with men (despite their increased age and frailty) reflects their longer life expectancy, smaller annular size (and less post-TAVR aortic regurgitation), more favorable LV reverse remodeling, or more likely, a combination of these factors remains to be established.
Collapse
Affiliation(s)
- Laura E Dobson
- Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds , Leeds, United Kingdom
| | - Timothy A Fairbairn
- Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds , Leeds, United Kingdom
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds , Leeds, United Kingdom
| | - John P Greenwood
- Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds , Leeds, United Kingdom
| |
Collapse
|
8
|
Yu L, Yang G, Weng X, Liang P, Li L, Li J, Fan Z, Tian W, Wu X, Xu H, Fang M, Ji Y, Li Y, Chen Q, Xu Y. Histone Methyltransferase SET1 Mediates Angiotensin II-Induced Endothelin-1 Transcription and Cardiac Hypertrophy in Mice. Arterioscler Thromb Vasc Biol 2015; 35:1207-17. [PMID: 25814673 DOI: 10.1161/atvbaha.115.305230] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 03/16/2015] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Endothelin-1 is a potent vasoconstrictor derived from vascular endothelium. Elevated endothelin-1 levels are observed in a host of cardiovascular pathologies including cardiomyopathy. The epigenetic mechanism responsible for endothelin-1 induction in these pathological processes remains elusive. APPROACH AND RESULTS We report here that induction of endothelin-1 expression in endothelial cells by angiotensin II (Ang II) was accompanied by the accumulation of histone H3K4 trimethylation, a preeminent histone modification for transcriptional activation, on the endothelin-1 promoter. In the meantime, Ang II stimulated the expression and the occupancy of Suv, Ez, and Trithorax domain 1 (SET1), a mammalian histone H3K4 trimethyltransferase, on the endothelin-1 promoter, both in vitro and in vivo. SET1 was recruited to the endothelin-1 promoter by activating protein 1 (c-Jun/c-Fos) and synergized with activating protein 1 to activate endothelin-1 transcription in response to Ang II treatment. Knockdown of SET1 in endothelial cells blocked Ang II-induced endothelin-1 synthesis and abrogated hypertrophy of cultured cardiomyocyte. Finally, endothelial-specific depletion of SET1 in mice attenuated Ang II-induced pathological hypertrophy and cardiac fibrosis. CONCLUSIONS Our data suggest that SET1 epigenetically activates endothelin-1 transcription in endothelial cells, thereby contributing to Ang II-induced cardiac hypertrophy. As such, screening of small-molecule compound that inhibits SET1 activity will likely offer a new therapeutic solution to the treatment of cardiomyopathy.
Collapse
Affiliation(s)
- Liming Yu
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Guang Yang
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Xinyu Weng
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Peng Liang
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Luyang Li
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Jianfei Li
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Zhiwen Fan
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Wenfang Tian
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Xiaoyan Wu
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Huihui Xu
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Minming Fang
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Yong Ji
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Yuehua Li
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Qi Chen
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.)
| | - Yong Xu
- From the Key Laboratory of Cardiovascular Disease and Department of Pathophysiology (L.Y., G.Y., X.W., P.L., L.L., J.L., Z.F., W.T., X.W., H.X., M.F., Y.J., Y.L., Q.C., Y.X.) and Laboratory Center for Basic Medical Sciences (X.W.), Nanjing Medical University, Nanjing, China; and Department of Surgery, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.).
| |
Collapse
|
9
|
Seksenyan A, Kadavallore A, Walts AE, de la Torre B, Berel D, Strom SP, Aliahmad P, Funari VA, Kaye J. TOX3 is expressed in mammary ER(+) epithelial cells and regulates ER target genes in luminal breast cancer. BMC Cancer 2015; 15:22. [PMID: 25632947 PMCID: PMC4324787 DOI: 10.1186/s12885-015-1018-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/13/2015] [Indexed: 01/13/2023] Open
Abstract
Background A breast cancer susceptibility locus has been mapped to the gene encoding TOX3. Little is known regarding the expression pattern or biological role of TOX3 in breast cancer or in the mammary gland. Here we analyzed TOX3 expression in murine and human mammary glands and in molecular subtypes of breast cancer, and assessed its ability to alter the biology of breast cancer cells. Methods We used a cell sorting strategy, followed by quantitative real-time PCR, to study TOX3 gene expression in the mouse mammary gland. To study the expression of this nuclear protein in human mammary glands and breast tumors, we generated a rabbit monoclonal antibody specific for human TOX3. In vitro studies were performed on MCF7, BT474 and MDA-MB-231 cell lines to study the effects of TOX3 modulation on gene expression in the context of breast cancer cells. Results We found TOX3 expression in estrogen receptor-positive mammary epithelial cells, including progenitor cells. A subset of breast tumors also highly expresses TOX3, with poor outcome associated with high expression of TOX3 in luminal B breast cancers. We also demonstrate the ability of TOX3 to alter gene expression in MCF7 luminal breast cancer cells, including cancer relevant genes TFF1 and CXCR4. Knockdown of TOX3 in a luminal B breast cancer cell line that highly expresses TOX3 is associated with slower growth. Surprisingly, TOX3 is also shown to regulate TFF1 in an estrogen-independent and tamoxifen-insensitive manner. Conclusions These results demonstrate that high expression of this protein likely plays a crucial role in breast cancer progression. This is in sharp contrast to previous studies that indicated breast cancer susceptibility is associated with lower expression of TOX3. Together, these results suggest two different roles for TOX3, one in the initiation of breast cancer, potentially related to expression of TOX3 in mammary epithelial cell progenitors, and another role for this nuclear protein in the progression of cancer. In addition, these results can begin to shed light on the reported association of TOX3 expression and breast cancer metastasis to the bone, and point to TOX3 as a novel regulator of estrogen receptor-mediated gene expression. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1018-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Akop Seksenyan
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Asha Kadavallore
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Ann E Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Brian de la Torre
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Dror Berel
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Center for Applied Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA.
| | - Samuel P Strom
- Genomics Core Facility, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Department of Pathology and Laboratory Medicine, University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Parinaz Aliahmad
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Vincent A Funari
- Genomics Core Facility, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Jonathan Kaye
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA. .,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Trefoil Factor 1 is involved in gastric cell copper homeostasis. Int J Biochem Cell Biol 2014; 59:30-40. [PMID: 25486181 DOI: 10.1016/j.biocel.2014.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/17/2014] [Accepted: 11/28/2014] [Indexed: 12/28/2022]
Abstract
Trefoil Factor 1 belongs to a group of small secreted proteins (the Trefoil Factor Family proteins), that are localized within the mucous granules and are expressed and secreted by epithelial cells that line mucous membranes. Trefoil factors are mainly expressed in the gastrointestinal tract, where they normally contribute to maintain the integrity of the mucosa. We recently demonstrated a selective binding ability of Trefoil Factor 1 for copper ions, through its carboxy-terminal tail, and we also observed that copper levels influence the equilibrium between the monomeric and homodimeric forms of Trefoil Factor 1, thus modulating its biological activity. Here we report that transcriptional regulation of Trefoil Factor 1 is also affected by copper levels, through the modulated binding of the copper-sensing transcription factor Sp1 onto the responsive elements present in the regulatory region of the gene. In addition we demonstrate that copper overload causes an accumulation of the trefoil protein in the Trans-Golgi Network and that Trefoil Factor 1 levels can influence copper excretion and copper related toxicity. These findings suggest that the protein might play a role in the overall complex mechanisms of copper homeostasis in the gastrointestinal tissues.
Collapse
|
11
|
Growth of poorly differentiated endometrial carcinoma is inhibited by combined action of medroxyprogesterone acetate and the Ras inhibitor Salirasib. Oncotarget 2014; 4:316-28. [PMID: 23530112 PMCID: PMC3712577 DOI: 10.18632/oncotarget.867] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Type 2 endometrial carcinoma (EC) is a poorly differentiated EC. Unlike type 1 EC, which responds to hormonal treatment (progestins), type 2 EC is refractory to hormonal treatment because of its low expression of active estrogen and progesterone receptors (ER, PR). The aim of this study was to develop a novel drug combination designed to treat these aggressive type 2 EC tumors without surgery and with fertility potential preserved. We examined the effects of combined treatment with the progestin medroxyprogesterone acetate (MPA) and the Ras inhibitor S-farnesylthiosalicylic acid (FTS; Salirasib). Because FTS can induce cell differentiation in tumor cells, we examined whether FTS could induce re-differentiation of type 2 EC cells, thereby sensitizing them to MPA. We found that FTS reduced Ras-GTP, phospho-Akt, and phospho-ERK, and that these reductions all correlated with a decrease in ERα phosphorylation. Combined treatment with FTS and MPA induced stronger reduction in USPC1 type 2 EC cell numbers than the reduction induced by either drug alone. MPA caused ERα degradation. Death of the cells was caused by MPA but not by FTS. The phosphorylated ERα induces gene transcription manifested by enhanced cell proliferation and survival. The combination of FTS and MPA, by reducing the mRNA expression of ERα-mediated genes (i.e. PR, c-fos and ps2/TFF1), inhibited tumor growth and enhanced the death of type 2 EC cells. These promising results might herald a novel treatment for the highly aggressive, incurable type 2 endometrial carcinoma.
Collapse
|
12
|
Markićević M, Džodić R, Buta M, Kanjer K, Mandušić V, Nešković-Konstantinović Z, Nikolić-Vukosavljević D. Trefoil factor 1 in early breast carcinoma: a potential indicator of clinical outcome during the first 3 years of follow-up. Int J Med Sci 2014; 11:663-73. [PMID: 24843314 PMCID: PMC4025164 DOI: 10.7150/ijms.8194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/15/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND A role of an estrogen-regulated, autocrine motogenic factor was assumed to be a major biological role of trefoil factor 1 (TFF1) in breast cancer. TFF1 is regarded as a predictive factor for positive response to endocrine therapy in breast cancer patients. The aim of our study was to examine TFF1 level distribution in breast carcinomas in order to distinguish estrogen-independent from estrogen-dependent TFF1 expression and to evaluate clinical usefulness of TFF1 status in early breast cancer during the first 3 years of follow-up. METHODS The study included 226 patients with primary operable invasive early breast carcinomas for whom an equal, a 3-year follow-up was conducted. TFF1 levels as well as estrogen receptor (ER) and progesterone receptor (PR) levels were measured in cytosolic extracts of tumor samples by immunoradiometric assay or by use of classical biochemical method, respectively. Non-parametric statistical tests were applied for data analyses. RESULTS Statistical analysis revealed that TFF1 levels were significantly higher in premenopausal patients (p=0.02), or in tumors with: lower histological grade (p<0.001), positive ER or PR status (p<0.001, in both cases). On the basis of TFF1 level distribution between ER-negative and ER-positive postmenopausal patients with tumors of different histological grade, 14 ng/mg was set as the cut-off value to distinguish estrogen-independent from estrogen-dependent TFF1 expression in breast cancer. Depending on menopausal and PR status, positive TFF1 status identified patients at opposite risk for relapse among ER-positive patients with grade II tumors. Among ER- and PR-positive premenopausal patients with grade II tumors, TFF1 status alone identified patients at opposite risk for relapse. CONCLUSIONS Determination of TFF1 status might identify patients at different risk for relapse and help in making decision on administering adjuvant therapy for early breast cancer patients during the first 3 years of follow-up.
Collapse
Affiliation(s)
- Milan Markićević
- 1. Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Radan Džodić
- 2. Surgical Oncology Clinic, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia; ; 3. University of Belgrade School of Medicine, Dr Subotića 8, 11000 Belgrade, Serbia
| | - Marko Buta
- 2. Surgical Oncology Clinic, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ksenija Kanjer
- 1. Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Vesna Mandušić
- 4. Vinča Institute of Nuclear Science, Mike Petrovića Alasa 12-14, 11000 Belgrade, Serbia
| | - Zora Nešković-Konstantinović
- 5. Clinic of Medical Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Dragica Nikolić-Vukosavljević
- 1. Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
13
|
RhoA and RhoC differentially modulate estrogen receptor α recruitment, transcriptional activities, and expression in breast cancer cells (MCF-7). J Cancer Res Clin Oncol 2013; 139:2079-88. [DOI: 10.1007/s00432-013-1533-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 09/24/2013] [Indexed: 01/14/2023]
|
14
|
Lang JD, Berry SM, Powers GL, Beebe DJ, Alarid ET. Hormonally responsive breast cancer cells in a microfluidic co-culture model as a sensor of microenvironmental activity. Integr Biol (Camb) 2013; 5:807-16. [PMID: 23559098 PMCID: PMC3648339 DOI: 10.1039/c3ib20265h] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Breast cancer cell growth and therapeutic response are manipulated extrinsically by microenvironment signals. Despite recognition of the importance of the microenvironment in a variety of tumor processes, predictive measures that incorporate the activity of the surrounding cellular environment are lacking. In contrast, tumor cell biomarkers are well established in the clinic. Expression of Estrogen Receptor-alpha (ERα) is the primary defining feature of hormonally responsive tumors and is the molecular target of therapy in the most commonly diagnosed molecular subtype of breast cancer. While a number of soluble factors have been implicated in ERα activation, the complexity of signaling between the cellular microenvironment and the cancer cell implies multivariate control. The cumulative impact of the microenvironment signaling, which we define as microenvironmental activity, is more difficult to predict than the sum of its parts. Here we tested the impact of an array of microenvironments on ERα signaling utilizing a microfluidic co-culture model. Quantitative immunofluorescence was employed to assess changes in ERα protein levels, combined with gene expression and phosphorylation status, as measures of activation. Analysis of microenvironment-induced growth under the same conditions revealed a previously undescribed correlation between growth and ERα protein down-regulation. These data suggest an expanded utility for the tumor biomarker ERα, in which the combination of dynamic regulation of ERα protein and growth in a breast cancer biosensor cell become a read-out of the microenvironmental activity.
Collapse
Affiliation(s)
- Jessica D Lang
- University of Wisconsin-Madison Carbone Comprehensive Cancer Center, Madison, WI 53705, USA
| | | | | | | | | |
Collapse
|
15
|
Hermani A, Shukla A, Medunjanin S, Werner H, Mayer D. Insulin-like growth factor binding protein-4 and -5 modulate ligand-dependent estrogen receptor-α activation in breast cancer cells in an IGF-independent manner. Cell Signal 2013; 25:1395-402. [PMID: 23499909 DOI: 10.1016/j.cellsig.2013.02.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 02/18/2013] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor binding proteins (IGFBPs) are modulators of numerous cellular processes including cell proliferation. Although IGFBPs classically act by sequestration of extracellular insulin-like growth factors (IGFs), thereby contributing to the fine-tuning of growth factor signals, IGF-independent actions of IGFBPs have also been described. In the breast, growth factor signaling in association with estradiol (E2)-stimulated estrogen receptor function is organized in a complex cross-talk. The importance of phosphatidylinositol 3-kinase/protein kinase B (Akt/PKB) pathway components for the E2-induced activation of estrogen receptor-alpha (ERα) is well accepted. Here we show that in the absence of IGFs, IGFBP-4 or IGFBP-5, either overexpressed in MCF-7 breast cancer cells or added exogenously, decreased the capability of E2 to induce ERα transcriptional activity. In addition, overexpression or addition of recombinant IGFBP-4 or IGFBP-5 resulted in reduction of E2-induced phosphorylation of Akt/PKB, GSK-3α/β and ERα in MCF-7 cells. The activation of the Akt/PKB-pathway describes a non-genomic effect of E2, which did not involve activation/phosphorylation of the IGF-I receptor (IGF-IR). Furthermore, knockdown of the IGF-IR did not affect the inhibition of E2-induced ERα phosphorylation by IGFBP-4 and 5. Moreover, IGFBP-4 and IGFBP-5 strongly decreased E2-triggered growth of MCF-7 cells. Our data suggest that IGFBPs interfere with the E2-induced activation of the Akt/PKB-pathway and prevent full hormone-dependent activation of ERα and breast cancer cell growth in an IGF- and IGF-IR-independent manner.
Collapse
Affiliation(s)
- Alexander Hermani
- Hormones and Signal Transduction Group, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
16
|
Médale-Giamarchi C, Lajoie-Mazenc I, Malissein E, Meunier E, Couderc B, Bergé Y, Filleron T, Keller L, Marty C, Lacroix-Triki M, Dalenc F, Doisneau-Sixou SF, Favre G. RhoB modifies estrogen responses in breast cancer cells by influencing expression of the estrogen receptor. Breast Cancer Res 2013; 15:R6. [PMID: 23339407 PMCID: PMC3672819 DOI: 10.1186/bcr3377] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 01/10/2013] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION RhoB has been reported to exert positive and negative effects on cancer pathophysiology but an understanding of its role in breast cancer remains incomplete. Analysis of data from the Oncomine database showed a positive correlation between RhoB expression and positivity for both estrogen receptor alpha (ERα) and progesterone receptor (PR). METHODS This finding was validated by our analysis of a tissue microarray constructed from a cohort of 113 patients and then investigated in human cell models. RESULTS We found that RhoB expression in tissue was strongly correlated with ERα and PR expression and inversely correlated with tumor grade, tumor size and count of mitosis. In human breast cancer cell lines, RhoB attenuation was associated with reduced expression of both ERα and PR, whereas elevation of RhoB was found to be associated with ERα overexpression. Mechanistic investigations suggested that RhoB modulates ERα expression, controlling both its protein and mRNA levels, and that RhoB modulates PR expression by accentuating the recruitment of ERα and other major co-regulators to the promoter of PR gene. A major consequence of RhoB modulation was that RhoB differentially regulated the proliferation of breast cancer cell lines. Interestingly, we documented crosstalk between RhoB and ERα, with estrogen treatment leading to RhoB activation. CONCLUSION Taken together, our findings offer evidence that in human breast cancer RhoB acts as a positive function to promote expression of ERα and PR in a manner correlated with cell proliferation.
Collapse
|
17
|
Insulin-like growth factor-I regulates GPER expression and function in cancer cells. Oncogene 2012; 32:678-88. [PMID: 22430216 DOI: 10.1038/onc.2012.97] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Functional cross talk between insulin-like growth factor-I (IGF-I) system and estrogen signaling has been largely reported, although the underlying molecular mechanisms remain to be fully elucidated. As GPR30/GPER mediates rapid cell responses to estrogens, we evaluated the potential of IGF-I to regulate GPER expression and function in estrogen receptor (ER)α-positive breast (MCF-7) and endometrial (Ishikawa) cancer cells. We found that IGF-I transactivates the GPER promoter sequence and upregulates GPER mRNA and protein levels in both cells types. Similar data were found, at least in part, in carcinoma-associated fibroblasts. The upregulation of GPER expression by IGF-I involved the IGF-IR/PKCδ/ERK/c-fos/AP1 transduction pathway and required ERα, as ascertained by specific pharmacological inhibitors and gene-silencing. In both MCF-7 and Ishikawa cancer cells, the IGF-I-dependent cell migration required GPER and its main target gene CTGF, whereas the IGF-I-induced proliferation required both GPER and cyclin D1. Our data demonstrate that the IGF-I system regulates GPER expression and function, triggering the activation of a signaling network that leads to the migration and proliferation of cancer cells.
Collapse
|
18
|
Bigsby RM, Caperell-Grant A. The role for estrogen receptor-alpha and prolactin receptor in sex-dependent DEN-induced liver tumorigenesis. Carcinogenesis 2011; 32:1162-6. [PMID: 21606321 DOI: 10.1093/carcin/bgr094] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mice treated neonatally with diethylnitrosamine (DEN) develop liver tumors in a male-dominant manner, reflecting the male bias in human hepatocellular carcinoma. Evidence suggests that estrogen, androgen, prolactin (PRL) and growth hormone (GH) modify liver tumorigenesis. We determined the roles of estrogen receptor-α (ERα) and prolactin receptor (PRLR) using receptor null mice, ERαKO (C57Bl/6J) and PRLR-KO (129Ola-X-C57BL/6), in the neonatal-DEN model of liver tumorigenesis. In both mouse strains, females had reduced tumorigenesis compared with males (P < 0.01), regardless of ERα or PRLR status. Tumorigenesis was not affected by ovariectomy in C57Bl/6J mice but it was increased by ovariectomy in the mixed strain, 129Ola-X-C57BL/6, regardless of PRLR status. ERαKO males had 47% fewer tumors than ERα wild-type males (P < 0.01). On the other hand, estradiol treatment protected against tumorigenesis in males only in the presence of ERα. As evidenced by liver gene expression, lack of ERα did not alter the pattern of GH secretion in males but resulted in the male GH pattern in females. These observations indicate that ERα is not required for lower tumorigenesis in females, but it is required for the protective effects of exogenously delivered estradiol. Unexpectedly, the results indicate that ERα plays a role in promotion of liver tumors in males. In addition, it can be concluded that sex differences in liver tumorigenesis cannot be explained by the sexually dimorphic pattern of GH secretion. The results also rule out PRL as the mediator of the protective effect of the ovaries.
Collapse
Affiliation(s)
- Robert M Bigsby
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 975 West Walnut Street, Indianapolis, IN 46202-5121, USA.
| | | |
Collapse
|
19
|
Ivanova MM, Luken KH, Zimmer AS, Lenzo FL, Smith RJ, Arteel MW, Kollenberg TJ, Mattingly KA, Klinge CM. Tamoxifen increases nuclear respiratory factor 1 transcription by activating estrogen receptor beta and AP-1 recruitment to adjacent promoter binding sites. FASEB J 2011; 25:1402-16. [PMID: 21233487 DOI: 10.1096/fj.10-169029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Little is known about endogenous estrogen receptor β (ERβ) gene targets in human breast cancer. We reported that estradiol (E(2)) induces nuclear respiratory factor-1 (NRF-1) transcription through ERα in MCF-7 breast cancer cells. Here we report that 4-hydroxytamoxifen (4-OHT), with an EC(50) of ~1.7 nM, increases NRF-1 expression by recruiting ERβ, cJun, cFos, CBP, and RNA polymerase II to and dismissing NCoR from the NRF1 promoter. Promoter deletion and transient transfection studies showed that the estrogen response element (ERE) is essential and that an adjacent AP-1 site contributes to maximal 4-OHT-induced NRF-1 transcription. siRNA knockdown of ERβ revealed that ERβ inhibits basal NRF-1 expression and is required for 4-OHT-induced NRF-1 transcription. An AP-1 inhibitor blocked 4-OHT-induced NRF-1 expression. The 4-OHT-induced increase in NRF-1 resulted in increased transcription of NRF-1 target CAPNS1 but not CYC1, CYC2, or TFAM despite increased NRF-1 coactivator PGC-1α protein. The absence of TFAM induction corresponds to a lack of Akt-dependent phosphorylation of NRF-1 with 4-OHT treatment. Overexpression of NRF-1 inhibited 4-OHT-induced apoptosis and siRNA knockdown of NRF-1 increased apoptosis, indicating an antiapoptotic role for NRF-1. Overall, NRF-1 expression and activity is regulated by 4-OHT via endogenous ERβ in MCF-7 cells.
Collapse
Affiliation(s)
- Margarita M Ivanova
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Dong S, Terasaka S, Kiyama R. Bisphenol A induces a rapid activation of Erk1/2 through GPR30 in human breast cancer cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2011; 159:212-218. [PMID: 20875696 DOI: 10.1016/j.envpol.2010.09.004] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 08/30/2010] [Accepted: 09/05/2010] [Indexed: 05/29/2023]
Abstract
Bisphenol A (BPA) has been considered as an endocrine disruptor due to its ability to interact with estrogen receptors (ERs). While G protein-coupled receptor 30 (GPR30) is a novel estrogen receptor, its role in BPA-induced activation of Erk1/2 remains unknown. Human breast cancer cell lines, MCF-7, MDA-MB-231 and SKBR3, were used as experimental models to discriminate between ERs-dependent, putative ERs-independent and/or GPR30-associated effects. BPA induced a rapid activation of Erk1/2 in both ERα/β-positive and negative breast cancer cells, and this effect was not blocked with an ER antagonist, ICI 182,780. A small interfering RNA assay revealed that the expression of GPR30 was necessary for BPA-induced activation of Erk1/2 and transcriptional regulation of c-fos. In addition, BPA regulates the expression of c-fos likely through an AP1-mediated pathway. As a conclusion, GPR30 plays an important role in the BPA-induced activation of Erk1/2 in a manner distinguishable from that in ERα-mediated signaling.
Collapse
Affiliation(s)
- S Dong
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8566, Japan; Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - S Terasaka
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8566, Japan
| | - R Kiyama
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8566, Japan.
| |
Collapse
|
21
|
Zhou L, Yan T, Jiang Y, Di G, Shen Z, Shao Z, Lu J. Prognostic and predictive value of TFF1 for adjuvant endocrine therapy in Chinese women with early ER positive breast cancer: comparing aromatase inhibitors with tamoxifen. Breast 2010; 20:15-20. [PMID: 20619646 DOI: 10.1016/j.breast.2010.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 06/01/2010] [Accepted: 06/12/2010] [Indexed: 01/15/2023] Open
Abstract
Factors that predict in favor of an aromatase inhibitors (AIs) over tamoxifen (TAM) in estrogen receptor (ER) breast cancer remains to be identified. We compared progesterone receptor (PR) and trefoil factor 1 (TTF1) status (+ve versus -ve) as predictive of superior effect of AI's over tamoxifen among a total of 1973 Chinese women with early ER+ breast cancer. The expression of TFF1 was independently associated with ER and PR. However, there was no correlation with TFF1 and HER-2 expression. Treatment effect was more pronounced in the ER+/TFF1+ postmenopausal patients with a hazard ratio favoring AIs (HR = 0.397, 95%CI 0.183-0.860), but not in the PR positive cohorts (HR = 0.466, 95%CI 0.186-1.164). We suggested that AIs was better than TAM especially in the postmenopausal patients with ER+/TFF1+ breast cancer; however the clinical application of this observation still requires further prospective studies.
Collapse
Affiliation(s)
- Liheng Zhou
- Department of Breast Surgery, Shanghai Cancer Hospital/Cancer Institute, Fudan University; Department of Oncology, Shanghai Medical College, Fudan University, 399 Ling-Ling Road, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Diotel N, Le Page Y, Mouriec K, Tong SK, Pellegrini E, Vaillant C, Anglade I, Brion F, Pakdel F, Chung BC, Kah O. Aromatase in the brain of teleost fish: expression, regulation and putative functions. Front Neuroendocrinol 2010; 31:172-92. [PMID: 20116395 DOI: 10.1016/j.yfrne.2010.01.003] [Citation(s) in RCA: 230] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 01/20/2010] [Accepted: 01/24/2010] [Indexed: 12/25/2022]
Abstract
Unlike that of mammals, the brain of teleost fish exhibits an intense aromatase activity due to the strong expression of one of two aromatase genes (aromatase A or cyp19a1a and aromatase B or cyp19a1b) that arose from a gene duplication event. In situ hybridization, immunohistochemistry and expression of GFP (green fluorescent protein) in transgenic tg(cyp19a1b-GFP) fish demonstrate that aromatase B is only expressed in radial glial cells (RGC) of adult fish. These cells persist throughout life and act as progenitors in the brain of both developing and adult fish. Although aromatase B-positive radial glial cells are most abundant in the preoptic area and the hypothalamus, they are observed throughout the entire central nervous system and spinal cord. In agreement with the fact that brain aromatase activity is correlated to sex steroid levels, the high expression of cyp19a1b is due to an auto-regulatory loop through which estrogens and aromatizable androgens up-regulate aromatase expression. This mechanism involves estrogen receptor binding on an estrogen response element located on the cyp19a1b promoter. Cell specificity is achieved by a mandatory cooperation between estrogen receptors and unidentified glial factors. Given the emerging roles of estrogens in neurogenesis, the unique feature of the adult fish brain suggests that, in addition to classical functions on brain sexual differentiation and sexual behaviour, aromatase expression in radial glial cells could be part of the mechanisms authorizing the maintenance of a high proliferative activity in the brain of fish.
Collapse
Affiliation(s)
- Nicolas Diotel
- Neurogenesis And OEstrogens, UMR CNRS 6026, IFR 140, Université de Rennes 1, Rennes, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Siewit CL, Gengler B, Vegas E, Puckett R, Louie MC. Cadmium promotes breast cancer cell proliferation by potentiating the interaction between ERalpha and c-Jun. Mol Endocrinol 2010; 24:981-92. [PMID: 20219890 DOI: 10.1210/me.2009-0410] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cadmium is an environmental contaminant that enters the body through diet or cigarette smoke. It affects multiple cellular processes, including cell proliferation, differentiation, and apoptosis. Recently, cadmium has been shown to function as an endocrine disruptor, to stimulate estrogen receptor alpha (ERalpha) activity and promote uterine and mammary gland growth in mice. Although cadmium exposure has been associated with the development of breast cancer, the mechanism of action of cadmium remains unclear. To address this deficit, we examined the effects of cadmium treatment on breast cancer cells. We found that ERalpha is required for both cadmium-induced cell growth and modulation of gene expression. We also determined that ERalpha translocates to the nucleus in response to cadmium exposure. Additionally, we provide evidence that cadmium potentiates the interaction between ERalpha and c-Jun and enhances recruitment of this transcription factor complex to the proximal promoters of cyclin D1 and c-myc, thus increasing their expression. This study provides a mechanistic link between cadmium exposure and ERalpha and demonstrates that cadmium plays an important role in the promotion of breast cancer.
Collapse
Affiliation(s)
- Christina L Siewit
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California 94901, USA
| | | | | | | | | |
Collapse
|
24
|
Piro M, Della Bona R, Abbate A, Biasucci LM, Crea F. Sex-Related Differences in Myocardial Remodeling. J Am Coll Cardiol 2010; 55:1057-65. [DOI: 10.1016/j.jacc.2009.09.065] [Citation(s) in RCA: 215] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 07/27/2009] [Accepted: 09/01/2009] [Indexed: 11/28/2022]
|
25
|
Hu DG, Mackenzie PI. Estrogen receptor alpha, fos-related antigen-2, and c-Jun coordinately regulate human UDP glucuronosyltransferase 2B15 and 2B17 expression in response to 17beta-estradiol in MCF-7 cells. Mol Pharmacol 2009; 76:425-39. [PMID: 19487245 DOI: 10.1124/mol.109.057380] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
UDP-glucuronosyltransferase 2B15 and 2B17 expression is up-regulated by 17beta-estradiol in MCF-7 breast cancer cells, as assessed by quantitative real-time polymerase chain reaction. Using 5'-deletion mapping and site-directed mutagenesis, we demonstrate that 17beta-estradiol activation of UGT2B15 gene transcription is mediated by a 282-base pair fragment positioned -454 to -172 nucleotides from the translation start site. This region contains two putative activator protein-1 (AP-1) elements, one imperfect estrogen response element (ERE), and two consensus ERE half-sites. We propose that these five sites act as an estrogen response unit (ERU), because mutation in any site reduces activation of the UGT2B15 promoter by 17beta-estradiol. Despite the presence of two AP-1 elements, the UGT2B15 promoter is not responsive to the AP-1 activator phorbol 12-myristate 13-acetate. Although electrophoretic mobility shift assays (EMSA) indicate that the AP-1 proteins c-Jun and Fos-related antigen 2 (Fra-2) bound to the distal AP-1 site, binding of Jun or Fos family members to the proximal AP-1 site was not detected by EMSA. Chromatin immunoprecipitation assays showed a 17beta-estradiol-induced recruitment of estrogen receptor (ER) alpha, c-Jun, and Fra-2 to the 282-bp ERU. The involvement of these three transcription factors in the stimulation of UGT2B15 gene expression by 17beta-estradiol was confirmed by siRNA silencing experiments. Mutagenesis and siRNA experiments indicate that UGT2B17 expression is also regulated by 17beta-estradiol via the ERU, which is fully conserved in both promoters. Because UGT2B15 and UGT2B17 inactivate steroid hormones by glucuronidation, the regulation of their genes by 17beta-estradiol may maintain steroid hormone homeostasis and prevent excessive estrogen signaling activity.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology, Flinders Medical Centre, Bedford Park, SA, Australia
| | | |
Collapse
|
26
|
Le Page Y, Menuet A, Kah O, Pakdel F. Characterization of a cis-acting element involved in cell-specific expression of the zebrafish brain aromatase gene. Mol Reprod Dev 2008; 75:1549-57. [PMID: 18288647 DOI: 10.1002/mrd.20892] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The cytochrome P450 Aromatase is the key enzyme catalyzing the conversion of androgens into estrogens. In zebrafish, the brain aromatase is encoded by cyp19b. Expression of cyp19b is restricted to radial glial cells bordering forebrain ventricles and is strongly stimulated by estrogens during development. At the promoter level, we have previously shown that an estrogen responsive element (ERE) is required for induction by estrogens. Here, we investigated the role of ERE flanking regions in the control of cell-specific expression. First, we show that a 20 bp length motif, named G x RE (glial x responsive element), acts in synergy with the ERE to mediate the estrogenic induction specifically in glial cells. Second, we demonstrate that, in vitro, this sequence binds factors exclusively present in glial or neuro-glial cells and is able to confer a glial specificity to an artificial estrogen-dependent gene. Taken together, these results contribute to the understanding of the molecular mechanisms allowing cyp19b regulation by estrogens and allowed to identify a promoter sequence involved in the strong estrogen inducibility of cyp19b which is specific for glial cells. The exceptional aromatase activity measured in the brain of teleost fish could rely on such mechanisms.
Collapse
Affiliation(s)
- Yann Le Page
- Université de Rennes 1, UMR CNRS 6026, Interactions Cellulaires et Moléculaires, Rennes, France
| | | | | | | |
Collapse
|
27
|
Fleury L, Gerus M, Lavigne AC, Richard-Foy H, Bystricky K. Eliminating epigenetic barriers induces transient hormone-regulated gene expression in estrogen receptor negative breast cancer cells. Oncogene 2008; 27:4075-85. [PMID: 18317449 DOI: 10.1038/onc.2008.41] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In breast cancer, approximately one-third of tumors express neither the estrogen receptor (ERalpha) nor estrogen-regulated genes such as the progesterone receptor gene (PR). Our study provides new insights into the mechanism allowing hormone-activated expression of ERalpha target genes silenced in ERalpha-negative mammary tumor cells. In cell lines derived from ERalpha-negative MDA-MB231 cells, stable expression of different levels of ERalpha from a transgene did not result in transcription of PR. A quantitative comparative analysis demonstrates that inhibiting DNA methyltransferases using 5-aza-2'-deoxycytidine or specific disruption of DNMT1 by small interfering RNAs and treatment with the histone-deacetylase inhibitor trichostatin A enabled ERalpha-mediated hormone-dependent expression of endogenous PR. We show that demethylation of a CpG island located in the first exon of PR was a prerequisite for ERalpha binding to these regulatory sequences. Although not a general requirement, DNA demethylation is also necessary for derepression of a subset of ERalpha target genes involved in tumorigenesis. PR transcription did not subsist 4 days after removal of the DNA methyltransferase blocking agents, suggesting that hormone-induced expression of ERalpha target genes in ERalpha-negative tumor cells is transient. Our observations support a model where an epigenetic mark confers stable silencing by precluding ERalpha access to promoters.
Collapse
Affiliation(s)
- L Fleury
- Laboratoire de Biologie Moléculaire Eucaryote, Université de Toulouse, Toulouse, France
| | | | | | | | | |
Collapse
|
28
|
Canettieri G, Franchi A, Guardia MD, Morantte I, Santaguida MG, Harney JW, Larsen PR, Centanni M. Activation of thyroid hormone is transcriptionally regulated by epidermal growth factor in human placenta-derived JEG3 cells. Endocrinology 2008; 149:695-702. [PMID: 17991726 PMCID: PMC2219305 DOI: 10.1210/en.2007-0779] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Accepted: 10/30/2007] [Indexed: 01/22/2023]
Abstract
Human type II deiodinase is a master regulator of thyroid hormone activation in several tissues. In placenta, type II deiodinase mRNA levels and enzymatic activity are elevated only during the first trimester of pregnancy and then progressively decline. During this early stage, mitogens such as epidermal growth factor (EGF) have been shown to promote the proliferation of the trophoblast by acting through multiple mechanisms. Here we show that EGF modulates transcription of human type II deiodinase gene (Dio2) through distinct signaling pathways, leading to the assembly of a heterogeneous transcription factor complex. Gene expression and deiodination assays have shown that EGF promptly induces a short-lived Dio2 mRNA and enzymatic activity. The induction is mediated by ERK and p38 kinases, as demonstrated by selective inhibition or overexpression of different mitogen-activated kinases. Reporter assays of mutant constructs indicate that EGF-induced transcriptional activity on Dio2 promoter is mediated by the cAMP response element (CRE) and does not involve the activating protein 1 site. With functional and biochemical approaches, we have demonstrated that the EGF stimulation culminates with the assembly and recruitment over the Dio2 CRE of a composite complex, which consists of c-Jun, c-Fos, and CRE-binding protein. These results further support the hypothesis that placental iodothyronine metabolism is critical during early pregnancy.
Collapse
Affiliation(s)
- Gianluca Canettieri
- Laboratory of Molecular Oncology, Dipartimento di Medicina Sperimentale, Viale Regina Elena, 324-00161, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|