1
|
Abbas MA, Al-Kabariti AY, Sutton C. Comprehensive understanding of the role of GPER in estrogen receptor-alpha negative breast cancer. J Steroid Biochem Mol Biol 2024; 241:106523. [PMID: 38636681 DOI: 10.1016/j.jsbmb.2024.106523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/01/2023] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
G protein-coupled estrogen receptor (GPER) plays a prominent role in facilitating the rapid, non-genomic signaling of estrogens in breast cancer cells. Herein, a comprehensive overview of the role of GPER in ER-ɑ-negative breast cancer is provided. Activation of GPER affected proliferation, metastasis and epithelial mesenchymal transition in ER-ɑ negative breast cancer cells. Clinical studies have demonstrated that GPER positivity was strongly correlated with larger tumor size and advanced clinical stage, suggesting that GPER/ERK signaling may play a role in promoting tumor progression. Strong evidence existed that environmental contaminants like bisphenol A have a carcinogenic potential mediated by GPER activation. The complexity of the cross talk between GPER and other receptors including ER-β, ER-α36, Estrogen-related receptor α (ERRα) and androgen receptor has been discussed. The potential utility of small molecules and phytoestrogens targeting GPER, adds valuable insights into its therapeutic potential. This review holds promises in advancing our understanding of GPER role in ER-ɑ-negative breast cancer. Overall, the consequences of GPER activation are still an area of active research and the implication are not entirely clear.
Collapse
Affiliation(s)
- Manal A Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Aya Y Al-Kabariti
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Chris Sutton
- School of Chemistry and Biosciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
2
|
Czaja A, Jiang AJ, Blanco MZ, Eremina OE, Zavaleta C. A Raman topography imaging method toward assisting surgical tumor resection. NPJ IMAGING 2024; 2:2. [PMID: 40051976 PMCID: PMC11884652 DOI: 10.1038/s44303-024-00006-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/10/2024] [Indexed: 03/09/2025]
Abstract
Achieving complete tumor resection upon initial surgical intervention can lead to better patient outcomes by making adjuvant treatments more efficacious and reducing the strain of repeat surgeries. Complete tumor resection can be difficult to confirm intraoperatively. Methods like touch preparation (TP) have been inconsistent for detecting residual malignant cell populations, and fatty specimens like breast cancer lumpectomies are too fatty to process for rapid histology. We propose a novel workflow of immunostaining and topographic surface imaging of freshly excised tissue to ensure complete resection using highly sensitive and spectrally separable surface-enhanced Raman scattering nanoparticles (SERS NPs) as the targeted contrast agent. Biomarker-targeting SERS NPs are ideal contrast agents for this application because their sensitivity enables rapid detection, and their narrow bands enable extensive intra-pixel multiplexing. The adaptive focus capabilities of an advanced Raman instrument, combined with our rotational accessory device for exposing each surface of the stained specimen to the objective lens, enable topographic mapping of complete excised specimen surfaces. A USB-controlled accessory for a Raman microscope was designed and fabricated to enable programmatic and precise angular manipulation of specimens in concert with instrument stage motions during whole-surface imaging. Specimens are affixed to the accessory on an anti-slip, sterilizable rod, and the tissue surface exposed to the instrument is adjusted on demand using a programmed rotating stepper motor. We demonstrate this topographic imaging strategy on a variety of phantoms and preclinical tissue specimens. The results show detail and texture in specimen surface topography, orientation of findings and navigability across surfaces, and extensive SERS NP multiplexing and linear quantitation capabilities under this new Raman topography imaging method. We demonstrate successful surface mapping and recognition of all 26 of our distinct SERS NP types along with effective deconvolution and localization of randomly assigned NP mixtures. Increasing NP concentrations were also quantitatively assessed and showed a linear correlation with Raman signal with an R2 coefficient of determination of 0.97. Detailed surface renderings color-encoded by unmixed SERS NP abundances show a path forward for content-rich, interactive surgical margin assessment.
Collapse
Affiliation(s)
- Alexander Czaja
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | - Alice J. Jiang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | - Matt Zacchary Blanco
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | - Olga E. Eremina
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | - Cristina Zavaleta
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
3
|
Al-Kabariti AY, Abbas MA. Progress in the Understanding of Estrogen Receptor Alpha Signaling in Triple-Negative Breast Cancer: Reactivation of Silenced ER-α and Signaling through ER-α36. Mol Cancer Res 2023; 21:1123-1138. [PMID: 37462782 DOI: 10.1158/1541-7786.mcr-23-0321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/21/2023] [Accepted: 07/14/2023] [Indexed: 11/02/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive tumor that accounts for approximately 15% of total breast cancer cases. It is characterized by poor prognosis and high rate of recurrence compared to other types of breast cancer. TNBC has a limited range of treatment options that include chemotherapy, surgery, and radiation due to the absence of estrogen receptor alpha (ER-α) rendering hormonal therapy ineffective. However, possible targets for improving the clinical outcomes in TNBC exist, such as targeting estrogen signaling through membranous ER-α36 and reactivating silenced ER-α. It has been shown that epigenetic drugs such as DNA methyltransferase and histone deacetylase inhibitors can restore the expression of ER-α. This reactivation of ER-α, presents a potential strategy to re-sensitize TNBC to hormonal therapy. Also, this review provides up-to-date information related to the direct involvement of miRNA in regulating the translation of ER-α mRNA. Specific epi-miRNAs can regulate ER-α expression indirectly by post-transcriptional targeting of mRNAs of enzymes that are involved in DNA methylation and histone deacetylation. Furthermore, ER-α36, an alternative splice variant of ER-α66, is highly expressed in ER-negative breast tumors and activates MAPK/ERK pathway, promoting cell proliferation, escaping apoptosis, and enhancing metastasis. In the future, these recent advances may be helpful for researchers working in the field to obtain novel treatment options for TNBC, utilizing epigenetic drugs and epi-miRNAs that regulate ER-α expression. Also, there is some evidence to suggest that drugs that decrease the expression of ER-α36 may be effective in treating TNBC.
Collapse
Affiliation(s)
- Aya Y Al-Kabariti
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
| | - Manal A Abbas
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| |
Collapse
|
4
|
Wong KY, Kong TH, Poon CCW, Yu W, Zhou L, Wong MS. Icariin, a phytoestrogen, exerts rapid estrogenic actions through crosstalk of estrogen receptors in osteoblasts. Phytother Res 2023; 37:4706-4721. [PMID: 37421324 DOI: 10.1002/ptr.7939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/10/2023]
Abstract
Icariin, a flavonoid glycoside derived from Epimedium brevicornum Maxim, exerts bone protective effects via estrogen receptors (ERs). This study aimed to investigate the role of ER-α66, ER-α36, and GPER in bone metabolism in osteoblasts following treatment with icariin. Human osteoblastic MG-63 cells and osteoblast-specific ER-α66 knockout mice were employed. The ERs crosstalk in the estrogenic action of icariin was evaluated in ER-α66-negative human embryonic kidney HEK293 cells. Icariin, like E2, regulated ER-α36 and GPER protein expression in osteoblasts by downregulating them and upregulating ER-α66. ER-α36 and GPER suppressed the actions of icariin and E2 in bone metabolism. However, the in vivo administration of E2 (2 mg/kg/day) or icariin (300 mg/kg/day) restored bone conditions in KO osteoblasts. ER-α36 and GPER expression increased significantly and rapidly activated and translocated in KO osteoblasts after treatment with E2 or icariin. ER-α36 overexpression in KO osteoblasts further promoted the OPG/RANKL ratio induced by E2 or icariin treatment. This study showed icariin and E2 elicit rapid estrogenic responses in bone through recruiting ER-α66, ER-α36, and GPER. Notably, in osteoblasts lacking ER-α66, ER-α36, and GPER mediate the estrogenic effects of icariin and E2, while in intact osteoblasts, ER-α36 and GPER act as negative regulators of ER-α66.
Collapse
Affiliation(s)
- Ka-Ying Wong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
| | - Tsz-Hung Kong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
| | - Christina Chui-Wa Poon
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
| | - Wenxuan Yu
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
| | - Liping Zhou
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
| | - Man-Sau Wong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, People's Republic of China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, People's Republic of China
| |
Collapse
|
5
|
Estrogen Receptor Alpha Splice Variants, Post-Translational Modifications, and Their Physiological Functions. Cells 2023; 12:cells12060895. [PMID: 36980236 PMCID: PMC10047206 DOI: 10.3390/cells12060895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
The importance of estrogenic signaling for a broad spectrum of biological processes, including reproduction, cancer development, energy metabolism, memory and learning, and so on, has been well documented. Among reported estrogen receptors, estrogen receptor alpha (ERα) has been known to be a major mediator of cellular estrogenic signaling. Accumulating evidence has shown that the regulations of ERα gene transcription, splicing, and expression across the tissues are highly complex. The ERα promoter region is composed of multiple leader exons and 5′-untranslated region (5′-UTR) exons. Differential splicing results in multiple ERα proteins with different molecular weights and functional domains. Furthermore, various post-translational modifications (PTMs) further impact ERα cellular localization, ligand affinity, and therefore functionality. These splicing isoforms and PTMs are differentially expressed in a tissue-specific manner, mediate certain aspects of ERα signaling, and may work even antagonistically against the full-length ERα. The fundamental understanding of the ERα splicing isoforms in normal physiology is limited and association studies of the splicing isoforms and the PTMs are scarce. This review aims to summarize the functional diversity of these ERα variants and the PTMs in normal physiological processes, particularly as studied in transgenic mouse models.
Collapse
|
6
|
Nagel A, Popeda M, Muchlinska A, Sadej R, Szade J, Zielinski J, Skokowski J, Niemira M, Kretowski A, Markiewicz A, Zaczek AJ. ERα36-High Cancer-Associated Fibroblasts as an Unfavorable Factor in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14082005. [PMID: 35454913 PMCID: PMC9024776 DOI: 10.3390/cancers14082005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Cancer-associated fibroblasts (CAFs) are the most abundant cell type in the tumor microenvironment (TME). Estrogen receptor alpha 36 (ERα36), the alternatively spliced variant of ERα, is described as an unfavorable factor when expressed in cancer cells. ERα can be expressed also in CAFs; however, the role of ERα36 in CAFs is unknown. Methods: Four CAF cultures were isolated from chemotherapy-naïve BC patients and characterized for ERα36 expression and the NanoString gene expression panel using isolated RNA. Conditioned media from CAF cultures were used to assess the influence of CAFs on triple-negative breast cancer (TNBC) cells using a matrigel 3D culture assay. Results: We found that ERα36high CAFs significantly induced the branching of TNBC cells in vitro (p < 0.001). They also produced a set of pro-tumorigenic cytokines compared to ERα36low CAFs, among which hepatocyte growth factor (HGF) was the main inducer of TNBC cell invasive phenotype in vitro (p < 0.001). Tumor stroma rich in ERα36high CAFs was correlated with high Ki67 expression (p = 0.041) and tumor-associated macrophages markers (CD68 and CD163, p = 0.041 for both). HGF was found to be an unfavorable prognostic factor in TCGA database analysis (p = 0.03 for DFS and p = 0.04 for OS). Conclusions: Breast cancer-associated fibroblasts represent distinct subtypes based on ERα36 expression. We propose that ERα36high CAFs could account for an unfavorable prognosis for TNBC patients.
Collapse
Affiliation(s)
- Anna Nagel
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.N.); (M.P.); (A.M.); (A.M.)
| | - Marta Popeda
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.N.); (M.P.); (A.M.); (A.M.)
| | - Anna Muchlinska
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.N.); (M.P.); (A.M.); (A.M.)
| | - Rafal Sadej
- Laboratory of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Jolanta Szade
- Department of Pathomorphology, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Jacek Zielinski
- Department of Surgical Oncology, Medical University of Gdansk, 80-211 Gdansk, Poland; (J.Z.); (J.S.)
| | - Jaroslaw Skokowski
- Department of Surgical Oncology, Medical University of Gdansk, 80-211 Gdansk, Poland; (J.Z.); (J.S.)
- Department of Medical Laboratory Diagnostics-Biobank Fahrenheit BBMRI.pl, Medical University of Gdansk, Debinki Street 7, 80-211 Gdansk, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, 15–276 Bialystok, Poland; (M.N.); (A.K.)
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, 15–276 Bialystok, Poland; (M.N.); (A.K.)
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.N.); (M.P.); (A.M.); (A.M.)
| | - Anna J. Zaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.N.); (M.P.); (A.M.); (A.M.)
- Correspondence: ; Tel.: +48–58-349–14-38
| |
Collapse
|
7
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
8
|
Xu Z, Zhao D, Zheng X, Huang B, Pan X, Xia X. Low concentrations of 17β-estradiol exacerbate tamoxifen resistance in breast cancer treatment through membrane estrogen receptor-mediated signaling pathways. ENVIRONMENTAL TOXICOLOGY 2022; 37:514-526. [PMID: 34821461 DOI: 10.1002/tox.23417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/09/2021] [Accepted: 11/14/2021] [Indexed: 06/13/2023]
Abstract
The present study aims to discover the influences of tamoxifen and 17β-estradiol (E2) on tamoxifen-resistant (TamR) patients in vitro. Herein, we established a stabilized TamR MCF-7 cell line at 1 μM via gradient concentrations of tamoxifen cultivation. The expression changes of four ER subtypes (ERα66, ERβ, ERα36 and GPR30) were found to bring about tamoxifen resistance. Moreover, the generation of tamoxifen resistance involved in apoptosis escape via a reactive oxygen species-regulated p53 signaling pathway. Interestingly, E2 at environmental concentrations (0.1-10 nM) could activate the expression of both ERα36 and GPR30, and then stimulate the phosphorylation of ERK1/2 and Akt, resulting in cell growth promotion. Cell migration and invasion promotion, apoptosis inhibition, and cell cycle G1-S progression are involved in such proliferative effects. Conversely, the application of specific antagonists of ERα36 and GPR30 could restore tamoxifen's sensitivity as well as partially offset E2-mediated proliferation. In short, overexpression of ERα36 and GPR30 not only ablate tamoxifen responsiveness but also could promote tumor progression of TamR breast cancer under estrogen conditions. These results provided novel insights into underlying mechanisms of tamoxifen resistance and the negative effects of steroid estrogens at environmental concentrations on TamR MCF-7 cells, thus generating new thoughts for future management of ER-positive breast cancer.
Collapse
Affiliation(s)
- Zhixiang Xu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Dimeng Zhao
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Xianyao Zheng
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Bin Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Xuejun Pan
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
9
|
Adlanmerini M, Fontaine C, Gourdy P, Arnal JF, Lenfant F. Segregation of nuclear and membrane-initiated actions of estrogen receptor using genetically modified animals and pharmacological tools. Mol Cell Endocrinol 2022; 539:111467. [PMID: 34626731 DOI: 10.1016/j.mce.2021.111467] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022]
Abstract
Estrogen receptor alpha (ERα) and beta (ERβ) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.
Collapse
Affiliation(s)
- Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France.
| |
Collapse
|
10
|
Zhao B, Ye X, Yang Y, Wang Y, Wang R, Pan X, Wang M. Knockdown of ER-α36 expression inhibits glioma proliferation, invasion and epithelial-to-mesenchymal transition. Anat Rec (Hoboken) 2021; 305:321-332. [PMID: 34331393 DOI: 10.1002/ar.24723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022]
Abstract
Estrogen receptor-α36 (ER-α36), a subtype of the estrogen receptor, is reported to play roles in tumorigenesis and tamoxifen resistance in several tumors, especially breast cancer. However, the role of ER-α36 in glioma proliferation and invasion remains unknown. Here, we explored the function of ER-α36 in glioma cells, using U87 and U251 cell lines. We found that ER-α36 was upregulated in glioma tissues compared to adjacent nontumor tissues. In U87 and U251 glioma cell lines, inhibition of ER-α36 expression by shRNA suppressed cell proliferation and invasion. In addition, the expression of an epithelial marker, ZO-1, was upregulated while that of one mesenchymal marker, N-cadherin, was downregulated with ER-α36 knockdown. We also found that inhibition of ER-α36 inactivated both PI3K/AKT and MEK/ERK signals. Taken together, these data indicated that overexpression of ER-α36 is associated with glioma proliferation and progression but that inhibition of ER-α36 leads to suppressed invasion and the epithelial-to-mesenchymal transition via PI3K/AKT and MEK/ERK pathway inactivation in glioma cells.
Collapse
Affiliation(s)
- Bowen Zhao
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, China
| | - Xiang Ye
- Department of Neurology, Cadre Clinic, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanyuan Yang
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, China.,Prenatal Diagnosis Center, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Yuxing Wang
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, China.,Prenatal Diagnosis Center, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Ru Wang
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, China.,Prenatal Diagnosis Center, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Xiaohua Pan
- Department of Breast and Thyroid surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Molin Wang
- Department of Genetics and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Basic Medical Science, Shandong University, Jinan, China.,Prenatal Diagnosis Center, Jinan Maternity and Child Care Hospital, Jinan, China
| |
Collapse
|
11
|
Wang Y, Pan X, Li Y, Wang R, Yang Y, Jiang B, Sun G, Shao C, Wang M, Gong Y. CUL4B renders breast cancer cells tamoxifen-resistant via miR-32-5p/ER-α36 axis. J Pathol 2021; 254:185-198. [PMID: 33638154 DOI: 10.1002/path.5657] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/28/2021] [Accepted: 02/24/2021] [Indexed: 12/24/2022]
Abstract
Tamoxifen (TAM) resistance is a significant clinical challenge in endocrine therapies for estrogen receptor (ER)-positive breast cancer patients. Cullin 4B (CUL4B), which acts as a scaffold protein in CUL4B-RING ubiquitin ligase complexes (CRL4B), is frequently overexpressed in cancer and represses tumor suppressors through diverse epigenetic mechanisms. However, the role and the underlying mechanisms of CUL4B in regulating drug resistance remain unknown. Here, we showed that CUL4B promotes TAM resistance in breast cancer cells through a miR-32-5p/ER-α36 axis. We found that upregulation of CUL4B correlated with decreased TAM sensitivity of breast cancer cells, and knockdown of CUL4B or expression of a dominant-negative CUL4B mutant restored the response to TAM in TAM-resistant MCF7-TAMR and T47D-TAMR cells. Mechanistically, we demonstrated that CUL4B renders breast cancer cells TAM-resistant by upregulating ER-α36 expression, which was mediated by downregulation of miR-32-5p. We further showed that CRL4B epigenetically represses the transcription of miR-32-5p by catalyzing monoubiquitination at H2AK119 and coordinating with PRC2 and HDAC complexes to promote trimethylation at H3K27 at the promoter of miR-32-5p. Pharmacologic or genetic inhibition of CRL4B/PRC2/HDAC complexes significantly increased TAM sensitivity in breast cancer cells in vitro and in vivo. Taken together, our findings thus establish a critical role for the CUL4B-miR-32-5p-ER-α36 axis in the regulation of TAM resistance and have important therapeutic implications for combined application of TAM and the inhibitors of CRL4B/PRC2/HDAC complex in breast cancer treatment. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yuxing Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xiaohua Pan
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, PR China
| | - Yanjun Li
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Ru Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yuanyuan Yang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Baichun Jiang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Gongping Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Changshun Shao
- State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, PR China
| | - Molin Wang
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education, Institute of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| |
Collapse
|
12
|
Costa AR, Lança de Oliveira M, Cruz I, Gonçalves I, Cascalheira JF, Santos CRA. The Sex Bias of Cancer. Trends Endocrinol Metab 2020; 31:785-799. [PMID: 32900596 DOI: 10.1016/j.tem.2020.07.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/07/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
Abstract
In hormone-dependent organs, sex hormones and dysregulated hormone signaling have well-documented roles in cancers of the breast and female reproductive organs including endometrium and ovary, as well as in prostate and testicular cancers in males. Strikingly, epidemiological data highlight significant differences between the sexes in the incidence of various cancers in nonreproductive organs, where the role of sex hormones has been less well studied. In an era when personalized medicine is gaining recognition, understanding the molecular, cellular, and biological differences between men and women is timely for developing more appropriate therapeutic interventions according to gender. We review evidence that sex hormones also shape many of the dysregulated cellular and molecular pathways that lead to cell proliferation and cancer in nonreproductive organs.
Collapse
Affiliation(s)
- Ana Raquel Costa
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
| | | | - Inês Cruz
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
| | - Isabel Gonçalves
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
| | - José Francisco Cascalheira
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal; Department of Chemistry, University of Beira Interior, Covilhã, Portugal
| | - Cecília R A Santos
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal.
| |
Collapse
|
13
|
Pal U, Ghosh S, Limaye AM. DNA methylation in the upstream CpG island of the GPER locus and its relationship with GPER expression in colon cancer cell lines. Mol Biol Rep 2020; 47:7547-7555. [PMID: 32936384 DOI: 10.1007/s11033-020-05817-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023]
Abstract
The G-protein coupled estrogen receptor (GPER), a proposed tumor suppressor, relays short-term non-genomic responses in target cells and tissues. It frequently undergoes down-modulation in primary tumors of the breast, ovary, and endometrium. Liu and co-workers recently reported loss of GPER expression in colorectal cancer and attributed it to DNA methylation-dependent silencing. We hypothesized that GPER expression is inversely correlated with methylation in the upstream CpG island (upCpGi) in the GPER locus. Methylation in the upCpGi was analysed by bisulfite sequencing and correlated with GPER expression in a panel of colon cancer cell lines. Eight downstream CpGs of the upCpGi was differentially methylated across the cell lines. Methylation in this differentially methylated region (DMR) correlated inversely with GPER expression. Two cell lines, namely SW620 and COLO-320DM, were compared in terms of their viability in response to varying concentrations of G1, a GPER specific agonist. SW-620 cells, which had the least methylated DMR and the highest level of GPER expression, showed significant loss of viability with 1 µM G1. COLO-320DM, which had the most methylated DMR and the lowest level of GPER expression, did not show a significant response to 1 µM G1. At 5 µM G1, SW620 cells showed a greater reduction in viability than COLO-320DM cells. DNA methylation in the DMR is inversely correlated with GPER expression. DNA methylation-dependent silencing of GPER may be, at least in part, the underlying reason behind the loss of estrogen's oncoprotective effect via GPER in the colon.
Collapse
Affiliation(s)
- Uttariya Pal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Sujasha Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Anil Mukund Limaye
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
14
|
Jusic A, Salgado-Somoza A, Paes AB, Stefanizzi FM, Martínez-Alarcón N, Pinet F, Martelli F, Devaux Y, Robinson EL, Novella S. Approaching Sex Differences in Cardiovascular Non-Coding RNA Research. Int J Mol Sci 2020; 21:E4890. [PMID: 32664454 PMCID: PMC7402336 DOI: 10.3390/ijms21144890] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is the biggest cause of sickness and mortality worldwide in both males and females. Clinical statistics demonstrate clear sex differences in risk, prevalence, mortality rates, and response to treatment for different entities of CVD. The reason for this remains poorly understood. Non-coding RNAs (ncRNAs) are emerging as key mediators and biomarkers of CVD. Similarly, current knowledge on differential regulation, expression, and pathology-associated function of ncRNAs between sexes is minimal. Here, we provide a state-of-the-art overview of what is known on sex differences in ncRNA research in CVD as well as discussing the contributing biological factors to this sex dimorphism including genetic and epigenetic factors and sex hormone regulation of transcription. We then focus on the experimental models of CVD and their use in translational ncRNA research in the cardiovascular field. In particular, we want to highlight the importance of considering sex of the cellular and pre-clinical models in clinical studies in ncRNA research and to carefully consider the appropriate experimental models most applicable to human patient populations. Moreover, we aim to identify sex-specific targets for treatment and diagnosis for the biggest socioeconomic health problem globally.
Collapse
Affiliation(s)
- Amela Jusic
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Tuzla, 75000 Tuzla, Bosnia and Herzegovina;
| | - Antonio Salgado-Somoza
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (A.S.-S.); (F.M.S.); (Y.D.)
| | - Ana B. Paes
- INCLIVA Biomedical Research Institute, Menéndez Pelayo 4 Accesorio, 46010 Valencia, Spain; (A.B.P.); (N.M.-A.)
| | - Francesca Maria Stefanizzi
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (A.S.-S.); (F.M.S.); (Y.D.)
| | - Núria Martínez-Alarcón
- INCLIVA Biomedical Research Institute, Menéndez Pelayo 4 Accesorio, 46010 Valencia, Spain; (A.B.P.); (N.M.-A.)
| | - Florence Pinet
- INSERM, CHU Lille, Institut Pasteur de Lille, University of Lille, U1167 F-59000 Lille, France;
| | - Fabio Martelli
- Molecular Cardiology Laboratory, Policlinico San Donato IRCCS, San Donato Milanese, 20097 Milan, Italy;
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445 Strassen, Luxembourg; (A.S.-S.); (F.M.S.); (Y.D.)
| | - Emma Louise Robinson
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Susana Novella
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, and INCLIVA Biomedical Research Institute, Menéndez Pelayo 4 Accesorio, 46010 Valencia, Spain
| |
Collapse
|
15
|
Thiebaut C, Konan HP, Guerquin MJ, Chesnel A, Livera G, Le Romancer M, Dumond H. The Role of ERα36 in Development and Tumor Malignancy. Int J Mol Sci 2020; 21:E4116. [PMID: 32526980 PMCID: PMC7312586 DOI: 10.3390/ijms21114116] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Estrogen nuclear receptors, represented by the canonical forms ERα66 and ERβ1, are the main mediators of the estrogen-dependent pathophysiology in mammals. However, numerous isoforms have been identified, stimulating unconventional estrogen response pathways leading to complex cellular and tissue responses. The estrogen receptor variant, ERα36, was cloned in 2005 and is mainly described in the literature to be involved in the progression of mammary tumors and in the acquired resistance to anti-estrogen drugs, such as tamoxifen. In this review, we will first specify the place that ERα36 currently occupies within the diversity of nuclear and membrane estrogen receptors. We will then report recent data on the impact of ERα36 expression and/or activity in normal breast and testicular cells, but also in different types of tumors including mammary tumors, highlighting why ERα36 can now be considered as a marker of malignancy. Finally, we will explain how studying the regulation of ERα36 expression could provide new clues to counteract resistance to cancer treatments in hormone-sensitive tumors.
Collapse
Affiliation(s)
- Charlène Thiebaut
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| | - Henri-Philippe Konan
- Université de Lyon, F-69000 Lyon, France; (H.-P.K.); (M.L.R.)
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Marie-Justine Guerquin
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiation, Université de Paris, Université Paris Saclay, CEA, F-92265 Fontenay aux Roses, France; (M.-J.G.); (G.L.)
| | - Amand Chesnel
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| | - Gabriel Livera
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiation, Université de Paris, Université Paris Saclay, CEA, F-92265 Fontenay aux Roses, France; (M.-J.G.); (G.L.)
| | - Muriel Le Romancer
- Université de Lyon, F-69000 Lyon, France; (H.-P.K.); (M.L.R.)
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Hélène Dumond
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (C.T.); (A.C.)
| |
Collapse
|
16
|
Wang Y, Lu H, Fang C, Xu J. Palmitoylation as a Signal for Delivery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:399-424. [DOI: 10.1007/978-981-15-3266-5_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Pagano MT, Ortona E, Dupuis ML. A Role for Estrogen Receptor alpha36 in Cancer Progression. Front Endocrinol (Lausanne) 2020; 11:506. [PMID: 32849292 PMCID: PMC7411082 DOI: 10.3389/fendo.2020.00506] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022] Open
Abstract
Estrogen receptor α (ERα) functions as a ligand dependent transcription factor that directly binds specific estrogen responsive elements, thus regulating the transcription of estrogen sensitive genes. ERα has also been shown to be associated with the plasma membrane (membrane associated ERα, mERα), concentrated in lipid rafts, plasma membrane microdomains with a distinct lipid composition, where it transduces membrane-initiated estrogen-dependent activation of the mitogen-activated protein (MAP) kinase signaling pathway. Two isoforms of ERα have been described: the "traditional" ERα66 (66 kDa) and a lower molecular weight variant: the ERα46 (46 kDa). More recently, a novel ERα variant with a molecular mass of 36 kDa (ERα36) has been discovered. Notably, ERα36 has been found expressed in different human tumor cells, including both ER- positive and ER- negative breast cancer cells. Estrogen signaling at the cell membrane via ERα36 appears as capable of activating multiple pathways of importance for cancer aggressiveness and metastatic potential. The presence of serum autoantibodies reacting with mERα (anti-ERα Abs) in a large percentage of patients with breast cancer has recently been reported by our group. These anti-ERα Abs seem to act as estrogen agonists rapidly triggering MAP kinase pathway activation thus inducing tumor cell proliferation and overcoming cell resistance to anti-estrogen drug tamoxifen. In this review, we describe the involvement of ERα36 in different tumors. We also report the potential pathogenetic activity of anti-ERα Abs and their implication in drug resistance.
Collapse
|
18
|
Thiebaut C, Chesnel A, Merlin JL, Chesnel M, Leroux A, Harlé A, Dumond H. Dual Epigenetic Regulation of ERα36 Expression in Breast Cancer Cells. Int J Mol Sci 2019; 20:ijms20112637. [PMID: 31146345 PMCID: PMC6600239 DOI: 10.3390/ijms20112637] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer remains the major cause of cancer-induced morbidity and mortality in women. Among the different molecular subtypes, luminal tumors yet considered of good prognosis often develop acquired resistance to endocrine therapy. Recently, misregulation of ERα36 was reported to play a crucial role in this process. High expression of this ERα isoform was associated to preneoplastic phenotype in mammary epithelial cells, disease progression, and enhanced resistance to therapeutic agents in breast tumors. In this study, we identified two mechanisms that could together contribute to ERα36 expression regulation. We first focused on hsa-miR-136-5p, an ERα36 3’UTR-targeting microRNA, the expression of which inversely correlated to the ERα36 one in breast cancer cells. Transfection of hsa-miR136-5p mimic in MCF-7 cells resulted in downregulation of ERα36. Moreover, the demethylating agent decitabine was able to stimulate hsa-miR-136-5p endogenous expression, thus indirectly decreasing ERα36 expression and counteracting tamoxifen-dependent stimulation. The methylation status of ERα36 promoter also directly modulated its expression level, as demonstrated after decitabine treatment of breast cancer cell and confirmed in a set of tumor samples. Taken together, these results open the way to a direct and an indirect ERα36 epigenetic modulation by decitabine as a promising clinical strategy to counteract acquired resistance to treatment and prevent relapse.
Collapse
Affiliation(s)
| | - Amand Chesnel
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France.
| | - Jean-Louis Merlin
- Université de Lorraine, CNRS, CRAN, Institut de Cancérologie de Lorraine, F-54000 Nancy, France.
| | - Maelle Chesnel
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France.
| | - Agnès Leroux
- Institut de Cancérologie de Lorraine, F-54000 Nancy, France.
| | - Alexandre Harlé
- Université de Lorraine, CNRS, CRAN, Institut de Cancérologie de Lorraine, F-54000 Nancy, France.
| | - Hélène Dumond
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France.
| |
Collapse
|
19
|
Nagel A, Szade J, Iliszko M, Elzanowska J, Welnicka-Jaskiewicz M, Skokowski J, Stasilojc G, Bigda J, Sadej R, Zaczek A, Markiewicz A. Clinical and Biological Significance of ESR1 Gene Alteration and Estrogen Receptors Isoforms Expression in Breast Cancer Patients. Int J Mol Sci 2019; 20:ijms20081881. [PMID: 30995757 PMCID: PMC6514554 DOI: 10.3390/ijms20081881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/20/2022] Open
Abstract
The amplification of estrogen receptor alpha (ERα) encoded by the ESR1 gene has been described as having a prognostic role in breast cancer patients. However, increased dosage of the ESR1 gene (tested by real-time PCR) is also observed in ER-negative breast cancers, which might suggest the expression of alternative isoforms of ERα (other than classical ERα of 66 kDa). In the current work, we have investigated the ESR1 gene dosage in 402 primary breast cancer patients as well as the expression of ERα isoforms—ERα66 and ERα36—on mRNA and protein levels. The obtained results were correlated with clinicopathological data of the patients. Results showed that increased ESR1 gene dosage is not related to ESR1 gene amplification measured by fluorescent in situ hybridization (FISH), but it correlates with the decreased expression of ERα66 isoform (p = 0.01). Interestingly, the short ER isoform ERα36 was expressed in samples with increased ESR1 gene dosage, suggesting that genomic aberration might influence the expression of that particular isoform. Similarly to ESR1 increased gene dosage, high ERα36 expression was linked with the decreased disease-free survival of the patients (p = 0.05), which was independent of the status of the classical ERα66 level in breast tumors.
Collapse
Affiliation(s)
- Anna Nagel
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-211 Gdansk, Poland.
| | - Jolanta Szade
- Department of Pathology, Medical University of Gdansk, 80-210 Gdansk, Poland.
| | - Mariola Iliszko
- Department of Biology and Genetics, Medical University of Gdansk, 80-211 Gdansk, Poland.
| | - Julia Elzanowska
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-211 Gdansk, Poland.
| | | | - Jaroslaw Skokowski
- Department of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland.
- Department of Medical Laboratory Diagnostics -Biobank, Medical University of Gdansk, Gdansk, 80-210 Gdansk, Poland.
- Biobanking and Biomolecular Resources Research Infrastructure (BBMRI.PL), 80-210 Gdansk, Poland.
| | - Grzegorz Stasilojc
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-211 Gdansk, Poland.
| | - Jacek Bigda
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-211 Gdansk, Poland.
| | - Rafal Sadej
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-211 Gdansk, Poland.
| | - Anna Zaczek
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-211 Gdansk, Poland.
| | - Aleksandra Markiewicz
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, 80-211 Gdansk, Poland.
| |
Collapse
|
20
|
Yan Y, Yu L, Castro L, Dixon D. ERα36, a variant of estrogen receptor α, is predominantly localized in mitochondria of human uterine smooth muscle and leiomyoma cells. PLoS One 2017; 12:e0186078. [PMID: 29020039 PMCID: PMC5636123 DOI: 10.1371/journal.pone.0186078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 09/25/2017] [Indexed: 01/22/2023] Open
Abstract
ERα36 is a naturally occurring, membrane-associated, isoform of estrogen receptor α. The expression of ERα36 is due to alternative splicing and different promoter usage. ERα36 is a dominant-negative effector of ERα66-mediated transactivational activities and has the potential to trigger membrane-initiated mitogenic, nongenomic, estrogen signaling; however, the subcellular localization of ERα36 remains controversial. To determine the cellular localization of ERα36 in estrogen-responsive human uterine smooth muscle (ht-UtSMC) and leiomyoma (fibroid; ht-UtLM) cells, we conducted systematic confocal microscopy and subcellular fractionation analysis using ERα36 antibodies. With Image J colocalizaton analysis plugin, confocal images were analyzed to obtain a Pearson’s Correlation Coefficient (PCC) to quantify signal colocalization of ERα36 with mitochondrial, endoplasmic reticulum, and cytoskeletal components in both cell lines. When cells were double-stained with an ERα36 antibody and a mitochondrial-specific dye, MitoTracker, the PCC for the two channel signals were both greater than 0.75, indicating strong correlation between ERα36 and mitochondrial signals in the two cell lines. A blocking peptide competition assay confirmed that the mitochondria-associated ERα36 signal detected by confocal analysis was specific for ERα36. In contrast, confocal images double-stained with an ERα36 antibody and endoplasmic reticulum or cytoskeletal markers, had PCCs that were all less than 0.4, indicating no or very weak signal correlation. Fractionation studies showed that ERα36 existed predominantly in membrane fractions, with minimal or undetected amounts in the cytosol, nuclear, chromatin, and cytoskeletal fractions. With isolated mitochondrial preparations, we confirmed that a known mitochondrial protein, prohibitin, was present in mitochondria, and by co-immunoprecipitation analysis that ERα36 was associated with prohibitin in ht-UtLM cells. The distinctive colocalization pattern of ERα36 with mitochondria in ht-UtSMC and ht-UtLM cells, and the association of ERα36 with a mitochondrial-specific protein suggest that ERα36 is localized primarily in mitochondria and may play a pivotal role in non-genomic signaling and mitochondrial functions.
Collapse
Affiliation(s)
- Yitang Yan
- Molecular Pathogenesis Group, National Toxicology Program Laboratory (NTPL), National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, North Carolina, United States of America
| | - Linda Yu
- Molecular Pathogenesis Group, National Toxicology Program Laboratory (NTPL), National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, North Carolina, United States of America
| | - Lysandra Castro
- Molecular Pathogenesis Group, National Toxicology Program Laboratory (NTPL), National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, North Carolina, United States of America
| | - Darlene Dixon
- Molecular Pathogenesis Group, National Toxicology Program Laboratory (NTPL), National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
21
|
Iyer JK, Kalra M, Kaul A, Payton ME, Kaul R. Estrogen receptor expression in chronic hepatitis C and hepatocellular carcinoma pathogenesis. World J Gastroenterol 2017; 23:6802-6816. [PMID: 29085224 PMCID: PMC5645614 DOI: 10.3748/wjg.v23.i37.6802] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/12/2017] [Accepted: 07/22/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate gender-specific liver estrogen receptor (ER) expression in normal subjects and patients with hepatitis C virus (HCV)-related cirrhosis and hepatocellular carcinoma (HCC). METHODS Liver tissues from normal donors and patients diagnosed with HCV-related cirrhosis and HCV-related HCC were obtained from the NIH Liver Tissue and Cell Distribution System. The expression of ER subtypes, ERα and ERβ, were evaluated by Western blotting and real-time RT-PCR. The subcellular distribution of ERα and ERβ was further determined in nuclear and cytoplasmic tissue lysates along with the expression of inflammatory [activated NF-κB and IκB-kinase (IKK)] and oncogenic (cyclin D1) markers by Western blotting and immunohistochemistry. The expression of ERα and ERβ was correlated with the expression of activated NF-κB, activated IKK and cyclin D1 by Spearman's correlation. RESULTS Both ER subtypes were expressed in normal livers but male livers showed significantly higher expression of ERα than females (P < 0.05). We observed significantly higher mRNA expression of ERα in HCV-related HCC liver tissues as compared to normals (P < 0.05) and ERβ in livers of HCV-related cirrhosis and HCV-related HCC subjects (P < 0.05). At the protein level, there was a significantly higher expression of nuclear ERα in livers of HCV-related HCC patients and nuclear ERβ in HCV-related cirrhosis patients as compared to normals (P < 0.05). Furthermore, we observed a significantly higher expression of phosphorylated NF-κB and cyclin D1 in diseased livers (P < 0.05). There was a positive correlation between the expression of nuclear ER subtypes and nuclear cyclin D1 and a negative correlation between cytoplasmic ER subtypes and cytoplasmic phosphorylated IKK in HCV-related HCC livers. These findings suggest that dysregulated expression of ER subtypes following chronic HCV-infection may contribute to the progression of HCV-related cirrhosis to HCV-related HCC. CONCLUSION Gender differences were observed in ERα expression in normal livers. Alterations in ER subtype expression observed in diseased livers may influence gender-related disparity in HCV-related pathogenesis.
Collapse
Affiliation(s)
- Janaki K Iyer
- Department of Biochemistry and Microbiology, Oklahoma State University-Center for Health Sciences, Tulsa, OK 74107, United States
- (Current Affiliation) Department of Natural Sciences, Northeastern State University, Tahlequah, OK 74464, United States
| | - Mamta Kalra
- Immatics US Inc, Houston, TX 77077, United States
| | - Anil Kaul
- Health Care Administration, Oklahoma State University-Center for Health Sciences, Tulsa, OK 74107, United States
| | - Mark E Payton
- Department of Statistics, Oklahoma State University, Stillwater, OK 74078, United States
| | - Rashmi Kaul
- Department of Biochemistry and Microbiology, Oklahoma State University-Center for Health Sciences, Tulsa, OK 74107, United States
| |
Collapse
|
22
|
Arnal JF, Lenfant F, Metivier R, Flouriot G, Henrion D, Adlanmerini M, Fontaine C, Gourdy P, Chambon P, Katzenellenbogen B, Katzenellenbogen J. Membrane and Nuclear Estrogen Receptor Alpha Actions: From Tissue Specificity to Medical Implications. Physiol Rev 2017; 97:1045-1087. [DOI: 10.1152/physrev.00024.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/19/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
Abstract
Estrogen receptor alpha (ERα) has been recognized now for several decades as playing a key role in reproduction and exerting functions in numerous nonreproductive tissues. In this review, we attempt to summarize the in vitro studies that are the basis of our current understanding of the mechanisms of action of ERα as a nuclear receptor and the key roles played by its two activation functions (AFs) in its transcriptional activities. We then depict the consequences of the selective inactivation of these AFs in mouse models, focusing on the prominent roles played by ERα in the reproductive tract and in the vascular system. Evidence has accumulated over the two last decades that ERα is also associated with the plasma membrane and activates non-nuclear signaling from this site. These rapid/nongenomic/membrane-initiated steroid signals (MISS) have been characterized in a variety of cell lines, and in particular in endothelial cells. The development of selective pharmacological tools that specifically activate MISS and the generation of mice expressing an ERα protein impeded for membrane localization have begun to unravel the physiological role of MISS in vivo. Finally, we discuss novel perspectives for the design of tissue-selective ER modulators based on the integration of the physiological and pathophysiological roles of MISS actions of estrogens.
Collapse
Affiliation(s)
- Jean-Francois Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Raphaël Metivier
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Gilles Flouriot
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Daniel Henrion
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Chambon
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Benita Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - John Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| |
Collapse
|
23
|
ER-α36 mediates estrogen-stimulated MAPK/ERK activation and regulates migration, invasion, proliferation in cervical cancer cells. Biochem Biophys Res Commun 2017; 487:625-632. [DOI: 10.1016/j.bbrc.2017.04.105] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 04/19/2017] [Indexed: 11/24/2022]
|
24
|
Shao X, Liu Y, Liu M, Wang Y, Yan L, Wang H, Ma L, Li YX, Zhao Y, Wang YL. Testosterone Represses Estrogen Signaling by Upregulating miR-22. Hypertension 2017; 69:721-730. [DOI: 10.1161/hypertensionaha.116.08468] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/10/2016] [Accepted: 12/26/2016] [Indexed: 12/11/2022]
Abstract
Preeclampsia, a multisystem syndrome occurring during mid- to late gestation in humans, is a leading cause of maternal and perinatal morbidity and mortality. Patients usually present with high circulating testosterone and reduced estradiol production, but the mechanisms remain unclear. Revealing the mechanism that modulating the imbalance of testosterone and estradiol in preeclampsia is of great value in understanding the cause of the disease. The placenta is the predominant source of steroid hormone production during gestation, and we observed markedly increased 17β-HSD3 (17β-hydroxysteroid dehydrogenase 3) levels and downregulated aromatase expression, the key enzymes responsible for synthesis of testosterone and estradiol, respectively, in preeclamptic placentas compared with controls. Furthermore, we found a significant upregulation of microRNA (miR)-22 in preeclamptic placentas. In a trophoblast cell line, JEG-3 cells, testosterone repressed the expression of aromatase and estrogen receptor α and the production of estradiol while promoting miR-22 expression. miR-22 directly targeted and inhibited estrogen receptor α expression while indirectly decreasing aromatase expression and estradiol production by interfering with estrogen receptor α signaling. Furthermore, inhibition of miR-22 expression significantly reversed the inhibitory effect of testosterone on de novo estradiol synthesis in human trophoblastic cells. The findings reveal a mechanism underlying the balanced production of androgen and estrogen modulated by miR-22 in the human placenta and provide new insights into the pathogenesis of preeclampsia from the aspect of endocrine regulation.
Collapse
Affiliation(s)
- Xuan Shao
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yanlei Liu
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Ming Liu
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yongqing Wang
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Liying Yan
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Hao Wang
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Liyang Ma
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yu-xia Li
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yangyu Zhao
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| | - Yan-ling Wang
- From the State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China (X.S., Y.L., M.L., H.W., L.M., Y.-x.L., Y.-l. W.); Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China (Y.W., L.Y., Y.Z.); and University of Chinese Academy of Sciences, Beijing, China (Y.L., H.W., L.M., Y.-l. W)
| |
Collapse
|
25
|
Thiebaut C, Chamard-Jovenin C, Chesnel A, Morel C, Djermoune EH, Boukhobza T, Dumond H. Mammary epithelial cell phenotype disruption in vitro and in vivo through ERalpha36 overexpression. PLoS One 2017; 12:e0173931. [PMID: 28301550 PMCID: PMC5354400 DOI: 10.1371/journal.pone.0173931] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/28/2017] [Indexed: 12/16/2022] Open
Abstract
Estrogen receptor alpha 36 (ERα36) is a variant of the canonical estrogen receptor alpha (ERα66), widely expressed in hormone sensitive cancer cells and whose high expression level correlates with a poor survival prognosis for breast cancer patients. While ERα36 activity have been related to breast cancer progression or acquired resistance to treatment, expression level and location of ERα36 are poorly documented in the normal mammary gland. Therefore, we explored the consequences of a ERα36 overexpression in vitro in MCF-10A normal mammary epithelial cells and in vivo in a unique model of MMTV-ERα36 transgenic mouse strain wherein ERα36 mRNA was specifically expressed in the mammary gland. By a combination of bioinformatics and computational analyses of microarray data, we identified hierarchical gene networks, downstream of ERα36 and modulated by the JAK2/STAT3 signaling pathway. Concomitantly, ERα36 overexpression lowered proliferation rate but enhanced migration potential and resistance to staurosporin-induced apoptosis of the MCF-10A cell line. In vivo, ERα36 expression led to duct epithelium thinning and disruption in adult but not in prepubescent mouse mammary gland. These phenotypes correlated with a loss of E-cadherin expression. Here, we show that an enhanced expression of ERα36 is sufficient, by itself, to disrupt normal breast epithelial phenotype in vivo and in vitro through a dominant-positive effect on nongenomic estrogen signaling pathways. These results also suggest that, in the presence of adult endogenous steroid levels, ERα36 overexpression in vivo contributes to alter mammary gland architecture which may support pre-neoplastic lesion and augment breast cancer risk.
Collapse
Affiliation(s)
- Charlène Thiebaut
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Clémence Chamard-Jovenin
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Amand Chesnel
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Chloé Morel
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - El-Hadi Djermoune
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Taha Boukhobza
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
| | - Hélène Dumond
- CNRS-Université de Lorraine, UMR 7039, Centre de Recherche en Automatique de Nancy, BP70239, Vandœuvre-lès-Nancy, France
- * E-mail:
| |
Collapse
|