1
|
Suganya S, Ashok BS, Ajith TA. A Recent Update on the Role of Estrogen and Progesterone in Alzheimer's Disease. Cell Biochem Funct 2024; 42:e70025. [PMID: 39663597 DOI: 10.1002/cbf.70025] [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: 08/12/2024] [Revised: 10/23/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD), one of the most prevalent neurodegenerative disease responsible for 60%-80% dementia cases globally. The disease is more prevalent among elder females. Female reproductive hormones are found to be essential for cellular activities in brain. The physiological role of neurotrophins and sex hormones in hippocampal region during neurogenesis and neuron differentiation was studied as well. In addition to triggering cellular pathways, estrogen and progesterone carry out a number of biological processes that lead to neuroprotection. They might have an impact on learning and memory. One of estrogen's modest antioxidant properties is its direct scavenging of free radicals. The neurotrophic effect of estrogen and progesterone can be explained by their ability to rise the expression of the brain-derived neurotrophic factor (BDNF) mRNA. Additionally, they have the ability to degrade beta-amyloid and stop inflammation, apoptotic neuronal cell death, and tau protein phosphorylation. To enhance their neuroprotective action, various cross-talking pathways in cells that are mediated by estrogen, progesterone, and BDNF receptors. This include signaling by mitogen-activated protein kinase/extracellular regulated kinase, phosphatidylinositol 3-kinase/protein kinase B, and phospholipase/protein kinase C. Clinical research to establish the significance of these substances are fragmented, despite publications claiming a lower prevalence of AD when medication is started before menopause. This review article emphasizes an update on the role of estrogen, and progesterone in AD.
Collapse
Affiliation(s)
- S Suganya
- Department of Biochemistry, Sri Ramachandra Medical College and Research Institute, Chennai, Tamil Nadu, India
| | - Ben Sundra Ashok
- Department of Biochemistry, Sri Ramachandra Medical College and Research Institute, Chennai, Tamil Nadu, India
| | - Thekkuttuparambil Ananthanarayanan Ajith
- Department of Biochemistry, Amala Institute of Medical Sciences, Thrissur, Kerala, India
- Amala Integrated Medical Research Department, Amala Institute of Medical Sciences, Thrissur, Kerala, India
| |
Collapse
|
2
|
Gandhi N, Omer S, Harrison RE. In Vitro Cell Culture Model for Osteoclast Activation during Estrogen Withdrawal. Int J Mol Sci 2024; 25:6134. [PMID: 38892322 PMCID: PMC11173070 DOI: 10.3390/ijms25116134] [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: 04/22/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Estrogen (17β-estradiol) deficiency post-menopause alters bone homeostasis whereby bone resorption by osteoclasts exceeds bone formation by osteoblasts, leading to osteoporosis in females. We established an in vitro model to examine the consequences of estrogen withdrawal (E2-WD) on osteoclasts derived from the mouse macrophage RAW 264.7 cell line and utilized it to investigate the mechanism behind the enhanced osteoclast activity post-menopause. We found that a greater population of osteoclasts that underwent E2-WD contained a podosome belt necessary for osteoclasts to adhere and resorb bone and possessed elevated resorptive activity compared to osteoclasts exposed to estrogen (E2) continuously. Our results show that compared to osteoclasts that received E2 continuously, those that underwent E2-WD had a faster rate of microtubule (MT) growth, reduced RhoA activation, and shorter podosome lifespan. Thus, altered podosome and MT dynamics induced by the withdrawal of estrogen supports podosome belt assembly/stability in osteoclasts, which may explain their enhanced bone resorption activity.
Collapse
Affiliation(s)
- Nisha Gandhi
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada;
| | - Safia Omer
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada;
| | - Rene E. Harrison
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada;
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada;
| |
Collapse
|
3
|
Chen G, Zhou T, Cao J, Li X, Zhu C, Wang L, Zou G, Liang H. Roles of estrogen receptors during sexual reversal in Pelodiscus sinensis. Mol Biol Rep 2024; 51:634. [PMID: 38727746 DOI: 10.1007/s11033-024-09482-w] [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: 11/28/2023] [Accepted: 03/26/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND The Chinese soft-shelled turtle, Pelodiscus sinensis, exhibits distinct sexual dimorphism, with the males growing faster and larger than the females. During breeding, all-male offspring can be obtained using 17β-estradiol (E2). However, the molecular mechanisms underlying E2-induced sexual reversal have not yet been elucidated. Previous studies have investigated the molecular sequence and expression characteristics of estrogen receptors (ERs). METHODS AND RESULTS In this study, primary liver cells and embryos of P. sinensis were treated with ER agonists or inhibitors. Cell incubation experiments revealed that nuclear ERs (nERs) were the main pathway for the transmission of estrogen signals. Our results showed that ERα agonist (ERα-ag) upregulated the expression of Rspo1, whereas ERα inhibitor (ERα-Inh) downregulated its expression. The expression of Dmrt1 was enhanced after ERα-Inh + G-ag treatment, indicating that the regulation of male genes may not act through a single estrogen receptor, but a combination of ERs. In embryos, only the ERα-ag remarkably promoted the expression levels of Rspo1, Wnt4, and β-catenin, whereas the ERα-Inh had a suppressive effect. Additionally, Dmrt1, Amh, and Sox9 expression levels were downregulated after ERβ inhibitor (ERβ-Inh) treatment. GPER agonist (G-ag) has a significant promotion effect on Rspo1, Wnt4, and β-catenin, while the inhibitor G-Inh does not affect male-related genes. CONCLUSIONS Overall, these results suggest that ERs play different roles during sexual reversal in P. sinensis and ERα may be the main carrier of estrogen-induced sexual reversal in P. sinensis. Further studies need to be performed to analyze the mechanism of ER action.
Collapse
Affiliation(s)
- Guobin Chen
- Yangtze River Fisheries Research Institute, Chinese Academy of Fisheries Science, Wuhan, 430223, China
| | - Tong Zhou
- Yangtze River Fisheries Research Institute, Chinese Academy of Fisheries Science, Wuhan, 430223, China
| | - Jizeng Cao
- Yangtze River Fisheries Research Institute, Chinese Academy of Fisheries Science, Wuhan, 430223, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiang Li
- Anhui Xijia Agricultural Development Co. Ltd, Bengbu, 233700, China
| | - Chengjun Zhu
- Anhui Xijia Agricultural Development Co. Ltd, Bengbu, 233700, China
| | - Long Wang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fisheries Science, Wuhan, 430223, China
| | - Guiwei Zou
- Yangtze River Fisheries Research Institute, Chinese Academy of Fisheries Science, Wuhan, 430223, China
| | - Hongwei Liang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fisheries Science, Wuhan, 430223, China.
| |
Collapse
|
4
|
Ben S, Li S, Gu D, Zhao L, Xu S, Ding Z, Chen S, Cheng Y, Xin J, Du M, Wang M. Benzo[a]pyrene exposure affects colorectal cancer susceptibility by regulating ERβ-mediated LINC02977 transcription. ENVIRONMENT INTERNATIONAL 2024; 184:108443. [PMID: 38277997 DOI: 10.1016/j.envint.2024.108443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/04/2023] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Abstract
Environmental pollutants known as polycyclic aromatic hydrocarbons (PAHs) are produced through the incomplete combustion of organic material. While PAHs have been investigated as genotoxicants, they can also operate through nongenotoxic pathways in estrogen-dependent malignancies, such as breast, cervical and ovarian cancer. However, whether PAHs induce colorectal cancer (CRC) risk through estrogenic effects is still illusive. Here, we systematically investigated the abnormal expression and activation of estrogen receptor beta (ERβ) regulated by PAHs in CRC as well as the underlying mechanisms of ERβ-mediated CRC risk. Based on the 300 plasma samples from CRC patients and healthy controls detected by GC-MS/MS, we found that the plasma concentrations of benzo[a]pyrene (BaP) were significantly higher in CRC cases than in healthy controls, with significant estrogenic effects. Moreover, histone deacetylase 2 (HDAC2)-induced deacetylation of the promoter decreases ERβ expression, which is associated with poor overall survival and advanced tumor stage. The study also revealed that BaP and estradiol (E2) had different carcinogenic effects, with BaP promoting cell proliferation and inhibiting apoptosis, while E2 had the opposite effects. Additionally, this study mapped ERβ genomic binding regions by performing ChIP-seq and ATAC-seq and identified genetic variants of rs1411680 and its high linkage disequilibrium SNP rs6477937, which were significantly associated with CRC risk through meta-analysis of two independent Chinese population genome-wide association studies comprising 2,248 cases and 3,173 controls and then validation in a large-scale European population. By integrating data from functional genomics, we validated the regulatory effect of rs6477937 as an ERβ binding-disrupting SNP that mediated allele-specific expression of LINC02977 in a long-range chromosomal interaction manner, which was found to be highly expressed in CRC tissues. Overall, this study suggests that the different active effects on ERβ by PAHs and endogenous E2 may play a crucial role in the development and progression of CRC and highlights the potential of targeting ERβ and its downstream targets for CRC prevention and treatment.
Collapse
Affiliation(s)
- Shuai Ben
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Shuwei Li
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Dongying Gu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210000, Jiangsu, China
| | - Lingyan Zhao
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shenya Xu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhutao Ding
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Silu Chen
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yifei Cheng
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Junyi Xin
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mulong Du
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China.
| |
Collapse
|
5
|
Birgersson M, Indukuri R, Lindquist L, Stepanauskaite L, Luo Q, Deng Q, Archer A, Williams C. Ovarian ERβ cistrome and transcriptome reveal chromatin interaction with LRH-1. BMC Biol 2023; 21:277. [PMID: 38031019 PMCID: PMC10688478 DOI: 10.1186/s12915-023-01773-1] [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: 03/03/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Estrogen receptor beta (ERβ, Esr2) plays a pivotal role in folliculogenesis and ovulation, yet its exact mechanism of action is mainly uncharacterized. RESULTS We here performed ERβ ChIP-sequencing of mouse ovaries followed by complementary RNA-sequencing of wild-type and ERβ knockout ovaries. By integrating the ERβ cistrome and transcriptome, we identified its direct target genes and enriched biological functions in the ovary. This demonstrated its strong impact on genes regulating organism development, cell migration, lipid metabolism, response to hypoxia, and response to estrogen. Cell-type deconvolution analysis of the bulk RNA-seq data revealed a decrease in luteal cells and an increased proportion of theca cells and a specific type of cumulus cells upon ERβ loss. Moreover, we identified a significant overlap with the gene regulatory network of liver receptor homolog 1 (LRH-1, Nr5a2) and showed that ERβ and LRH-1 extensively bound to the same chromatin locations in granulosa cells. Using ChIP-reChIP, we corroborated simultaneous ERβ and LRH-1 co-binding at the ERβ-repressed gene Greb1 but not at the ERβ-upregulated genes Cyp11a1 and Fkbp5. Transactivation assay experimentation further showed that ERβ and LRH-1 can inhibit their respective transcriptional activity at classical response elements. CONCLUSIONS By characterizing the genome-wide endogenous ERβ chromatin binding, gene regulations, and extensive crosstalk between ERβ and LRH-1, along with experimental corroborations, our data offer genome-wide mechanistic underpinnings of ovarian physiology and fertility.
Collapse
Affiliation(s)
- Madeleine Birgersson
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Rajitha Indukuri
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
| | - Linnéa Lindquist
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Lina Stepanauskaite
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Qing Luo
- Department of Physiology and Pharmacology, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Amena Archer
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden
| | - Cecilia Williams
- Science for Life Laboratory (SciLifeLab), Department of Protein Science, KTH Royal Institute of Technology, 171 21, Solna, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83, Huddinge, Sweden.
| |
Collapse
|
6
|
Yan S, Wang J, Chen H, Zhang D, Imam M. Divergent features of ERβ isoforms in triple negative breast cancer: progress and implications for further research. Front Cell Dev Biol 2023; 11:1240386. [PMID: 37936981 PMCID: PMC10626554 DOI: 10.3389/fcell.2023.1240386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
Estrogen receptor β (ERβ) was discovered more than 20 years ago. However, the extent and role of ERβ expression in breast cancer remain controversial, especially in the context of triple-negative breast cancer (TNBC). ERβ exists as multiple isoforms, and a series of studies has revealed an inconsistent role of ERβ isoforms in TNBC. Our recent results demonstrated contrasting functions of ERβ1 and ERβ2/β5 in TNBC. Additional research should be conducted to explore the functions of individual ERβ isoforms and develop targeted drugs according to the relevant mechanisms. Consequently, a systematic review of ERβ isoforms is necessary. In this review, we overview the structure of ERβ isoforms and detail what is known about the function of ERβ isoforms in normal mammary tissue and breast cancer. Moreover, this review highlights the divergent features of ERβ isoforms in TNBC. This review also provides insights into the implications of targeting ERβ isoforms for clinical treatment. In conclusion, this review provides a framework delineating the roles and mechanisms of different ERβ isoforms in TNBC and sheds light on future directions for basic and clinical research.
Collapse
Affiliation(s)
- Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | | | | | | | | |
Collapse
|
7
|
Repeated exposure to 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP) accelerates ligand-independent activation of estrogen receptors in long-term estradiol-deprived MCF-7 cells. Toxicol Lett 2023; 378:31-38. [PMID: 36863540 DOI: 10.1016/j.toxlet.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/02/2023]
Abstract
It was previously identified that there may be an active metabolite of bisphenol A (BPA), 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP). An in vitro system was developed to detect MBP toxicity to the Michigan Cancer Foundation-7 (MCF-7) cells that had been repeatedly exposed to a low dose of the metabolite. MBP profoundly activated estrogen receptor (ER)-dependent transcription as a ligand, with an EC50 of 2.8 nM. Women are continuously exposed to numerous estrogenic environmental chemicals; but their susceptibility to these chemicals may be significantly altered after menopause. Long-term estrogen-deprived (LTED) cells, which display ligand-independent ER activation, are a postmenopausal breast cancer model derived from MCF-7 cells. In this study, we investigated the estrogenic effects of MBP on LTED cells in a repeated exposure in vitro model. The results suggest that i) nanomolar levels of MBP reciprocally disrupt the balanced expression of ERα and ERβ proteins, leading to the dominant expression of ERβ, ii) MBP stimulates ERs-mediated transcription without acting as an ERβ ligand, and iii) MBP utilizes mitogen-activated protein kinase and phosphatidylinositol-3 kinase signaling to evoke its estrogenic action. Moreover, the repeated exposure strategy was effective for detecting low-dose estrogenic-like effects caused by MBP in LTED cells.
Collapse
|
8
|
Cai Y, Shen X, Lu L, Yan H, Huang H, Gaule P, Muca E, Theriot CM, Rattray Z, Rattray NJW, Lu J, Ahuja N, Zhang Y, Paty PB, Khan SA, Johnson CH. Bile acid distributions, sex-specificity, and prognosis in colorectal cancer. Biol Sex Differ 2022; 13:61. [PMID: 36274154 PMCID: PMC9590160 DOI: 10.1186/s13293-022-00473-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bile acids are known to be genotoxic and contribute to colorectal cancer (CRC). However, the link between CRC tumor bile acids to tumor location, patient sex, microbiome, immune-regulatory cells, and prognosis is not clear. METHODS We conducted bile acid analysis using targeted liquid chromatography-mass spectrometry (LC-MS) on tumor tissues from CRC patients (n = 228) with survival analysis. We performed quantitative immunofluorescence (QIF) on tumors to examine immune cells. RESULTS Twelve of the bile acids were significantly higher in right-sided colon tumors compared to left-sided colon tumors. Furthermore, in male patients, right-sided colon tumors had elevated secondary bile acids (deoxycholic acid, lithocholic acid, ursodeoxycholic acid) compared to left-sided colon tumors, but this difference between tumors by location was not observed in females. A high ratio of glycoursodeoxycholic to ursodeoxycholic was associated with 5-year overall survival (HR = 3.76, 95% CI = 1.17 to 12.1, P = 0.026), and a high ratio of glycochenodeoxycholic acid to chenodeoxycholic acid was associated with 5-year recurrence-free survival (HR = 3.61, 95% CI = 1.10 to 11.84, P = 0.034). We also show correlation between these bile acids and FoxP3 + T regulatory cells. CONCLUSIONS This study revealed that the distribution of bile acid abundances in colon cancer patients is tumor location-, age- and sex-specific, and are linked to patient prognosis. This study provides new implications for targeting bile acid metabolism, microbiome, and immune responses for colon cancer patients by taking into account primary tumor location and sex.
Collapse
Affiliation(s)
- Yuping Cai
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510 USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510 USA
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT 06510 USA
| | - Hong Yan
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510 USA
| | - Huang Huang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510 USA
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Patricia Gaule
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Engjel Muca
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | | | - Zahra Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE UK
| | - Nicholas J. W. Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE UK
| | - Jun Lu
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520 USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Nita Ahuja
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Surgery, Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Yawei Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510 USA
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Philip B. Paty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Sajid A. Khan
- Department of Surgery, Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510 USA
| |
Collapse
|
9
|
Bonfim Neto AP, Cardoso APMM, Silva RDS, Sousa LMMDC, Giometti IC, Binelli M, Bauersachs S, Kowalewski MP, Papa PDC. An approach to uncover the relationship between 17b-estradiol and ESR1/ESR2 ratio in the regulation of canine corpus luteum. Front Vet Sci 2022; 9:885257. [PMID: 35982918 PMCID: PMC9378837 DOI: 10.3389/fvets.2022.885257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022] Open
Abstract
The canine corpus luteum (CL) is able to synthetise, activate and deactivate 17b-estradiol (E2) and also expresses nuclear estrogen receptors in a time-dependent manner during diestrus. Nevertheless, we are still missing a better comprehension of E2 functions in the canine CL, especially regarding the specific roles of estrogen receptor alpha (ERa) and ERb, encoded by ESR1 and 2, respectively. For that purpose, we analyzed transcriptomic data of canine non-pregnant CL collected on days 10, 20, 30, 40, 50 and 60 of diestrus and searched for differentially expressed genes (DEG) containing predicted transcription factor binding sites (TFBS) for ESR1 or ESR2. Based on biological functions of DEG presenting TFBS, expression of select transcripts and corresponding proteins was assessed. Additionally, luteal cells were collected across specific time points during diestrus and specificity of E2 responses was tested using ERa and/or ERb inhibitors. Bioinformatic analyses revealed 517 DEGs containing TFBS, from which 67 for both receptors. In general, abundance of predicted ESR1 targets was greater in the beginning, while abundance of ESR2 targets was greater in the end of diestrus. ESR1/ESR2 ratio shifted from an increasing to a decreasing pattern from day 30 to 40 post ovulation. Specific receptor inhibition suggested an ERa-mediated positive regulation of CL function at the beginning of diestrus and an ERb-mediated effect contributing to luteal regression. In conclusion, our data points toward a broad spectrum of action of E2 and its nuclear receptors, which can also act as transcription factors for other genes regulating canine CL function.
Collapse
Affiliation(s)
| | | | - Renata dos Santos Silva
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Ines Cristina Giometti
- Faculty of Veterinary Medicine, University of Western São Paulo, Presidente Prudente, Brazil
| | - Mario Binelli
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Stefan Bauersachs
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | - Paula de Carvalho Papa
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- *Correspondence: Paula de Carvalho Papa
| |
Collapse
|
10
|
Estrogen Receptor β (ESR2) Transcriptome and Chromatin Binding in a Mantle Cell Lymphoma Tumor Model Reveal the Tumor-Suppressing Mechanisms of Estrogens. Cancers (Basel) 2022; 14:cancers14133098. [PMID: 35804870 PMCID: PMC9264873 DOI: 10.3390/cancers14133098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Mantle cell lymphoma (MCL) is much more common in males than in females. The reason for this is not clear, but research has indicated that the female sex hormones, estrogens, have a protective effect on MCL development. To study this further, mice were transplanted with MCL cells and treated with an estrogen that selectively activates ESR2, the main nuclear estrogen receptor in lymphoma cells. The activation of ESR2 resulted in reduced MCL tumor growth of MCL tumors that were both sensitive and resistant to a newly developed drug (ibrutinib). The mechanism for this effect was investigated by analyzing gene expression and ESR2 binding to target genes. The results show that the affected genes were enriched in several malignancy-related biological processes, including MCL. Furthermore, the results suggested an interplay between the lymphoma cells and the tumor microenvironment in response to ESR2 activation. Altogether, the results clarify the mechanisms of ESR2-mediated MCL growth impairment by estrogens and provide a possible explanation for the sex difference in incidence. Furthermore, targeting ESR2 may be an option when considering the treatment of MCL. Abstract Mantle cell lymphoma (MCL) is a non-Hodgkin lymphoma with one of the highest male-to-female incidence ratios. The reason for this is not clear, but epidemiological as well as experimental data have suggested a role for estrogens, particularly acting through estrogen receptor β (ESR2). To study the ESR2 effects on MCL progression, MCL cells sensitive and resistant to the Bruton tyrosine kinase inhibitor ibrutinib were grafted to mice and treated with the ESR2-selective agonist diarylpropionitrile (DPN). The results showed that the DPN treatment of mice grafted with both ibrutinib-sensitive and -resistant MCL tumors resulted in impaired tumor progression. To identify the signaling pathways involved in the impaired tumor progression following ESR2 agonist treatment, the transcriptome and ESR2 binding to target genes were investigated by genome-wide chromatin immunoprecipitation in Granta-519 MCL tumors. DPN-regulated genes were enriched in several biological processes that included cell–cell adhesion, endothelial–mesenchymal transition, nuclear factor-kappaB signaling, vasculogenesis, lymphocyte proliferation, and apoptosis. In addition, downregulation of individual genes, such as SOX11 and MALAT1, that play a role in MCL progression was also observed. Furthermore, the data suggested an interplay between the lymphoma cells and the tumor microenvironment in response to the ESR2 agonist. In conclusion, the results clarify the mechanisms by which estrogens, via ESR2, impair MCL tumor progression and provide a possible explanation for the sex-dependent difference in incidence. Furthermore, targeting ESR2 with a selective agonist may be an additional option when considering the treatment of both ibrutinib-sensitive and -resistant MCL tumors.
Collapse
|
11
|
Chauvin S, Cohen-Tannoudji J, Guigon CJ. Estradiol Signaling at the Heart of Folliculogenesis: Its Potential Deregulation in Human Ovarian Pathologies. Int J Mol Sci 2022; 23:ijms23010512. [PMID: 35008938 PMCID: PMC8745567 DOI: 10.3390/ijms23010512] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/26/2023] Open
Abstract
Estradiol (E2) is a major hormone controlling women fertility, in particular folliculogenesis. This steroid, which is locally produced by granulosa cells (GC) within ovarian follicles, controls the development and selection of dominant preovulatory follicles. E2 effects rely on a complex set of nuclear and extra-nuclear signal transduction pathways principally triggered by its nuclear receptors, ERα and ERβ. These transcription factors are differentially expressed within follicles, with ERβ being the predominant ER in GC. Several ERβ splice isoforms have been identified and display specific structural features, which greatly complicates the nature of ERβ-mediated E2 signaling. This review aims at providing a concise overview of the main actions of E2 during follicular growth, maturation, and selection in human. It also describes the current understanding of the various roles of ERβ splice isoforms, especially their influence on cell fate. We finally discuss how E2 signaling deregulation could participate in two ovarian pathogeneses characterized by either a follicular arrest, as in polycystic ovary syndrome, or an excess of GC survival and proliferation, leading to granulosa cell tumors. This review emphasizes the need for further research to better understand the molecular basis of E2 signaling throughout folliculogenesis and to improve the efficiency of ovarian-related disease therapies.
Collapse
|
12
|
Song D, He H, Indukuri R, Huang Z, Stepanauskaite L, Sinha I, Haldosén LA, Zhao C, Williams C. ERα and ERβ Homodimers in the Same Cellular Context Regulate Distinct Transcriptomes and Functions. Front Endocrinol (Lausanne) 2022; 13:930227. [PMID: 35872983 PMCID: PMC9299245 DOI: 10.3389/fendo.2022.930227] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
The two estrogen receptors ERα and ERβ are nuclear receptors that bind estrogen (E2) and function as ligand-inducible transcription factors. They are homologues and can form dimers with each other and bind to the same estrogen-response element motifs in the DNA. ERα drives breast cancer growth whereas ERβ has been reported to be anti-proliferative. However, they are rarely expressed in the same cells, and it is not fully investigated to which extent their functions are different because of inherent differences or because of different cellular context. To dissect their similarities and differences, we here generated a novel estrogen-dependent cell model where ERα homodimers can be directly compared to ERβ homodimers within the identical cellular context. By using CRISPR-cas9 to delete ERα in breast cancer MCF7 cells with Tet-Off-inducible ERβ expression, we generated MCF7 cells that express ERβ but not ERα. MCF7 (ERβ only) cells exhibited regulation of estrogen-responsive targets in a ligand-dependent manner. We demonstrated that either ER was required for MCF7 proliferation, but while E2 increased proliferation via ERα, it reduced proliferation through a G2/M arrest via ERβ. The two ERs also impacted migration differently. In absence of ligand, ERβ increased migration, but upon E2 treatment, ERβ reduced migration. E2 via ERα, on the other hand, had no significant impact on migration. RNA sequencing revealed that E2 regulated a transcriptome of around 800 genes via each receptor, but over half were specific for either ERα or ERβ (417 and 503 genes, respectively). Functional gene ontology enrichment analysis reinforced that E2 regulated cell proliferation in opposite directions depending on the ER, and that ERβ specifically impacted extracellular matrix organization. We corroborated that ERβ bound to cis-regulatory chromatin of its unique proposed migration-related direct targets ANXA9 and TFAP2C. In conclusion, we demonstrate that within the same cellular context, the two ERs regulate cell proliferation in the opposite manner, impact migration differently, and each receptor also regulates a distinct set of target genes in response to E2. The developed cell model provides a novel and valuable resource to further complement the mechanistic understanding of the two different ER isoforms.
Collapse
Affiliation(s)
- Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Maternal and Child Health Care Hospital of Shandong Province, Jinan, China
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Huan He
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- School of Public Health, Jilin University, Changchun, China
| | - Rajitha Indukuri
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
| | - Zhiqiang Huang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Lina Stepanauskaite
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
| | - Indranil Sinha
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Arne Haldosén
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Cecilia Williams
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
- *Correspondence: Cecilia Williams,
| |
Collapse
|
13
|
Abstract
To identify regulators of triple-negative breast cancer (TNBC), gene expression profiles of malignant parts of TNBC (mTNBC) and normal adjacent (nadj) parts of the same breasts have been compared. We are interested in the roles of estrogen receptor β (ERβ) and the cytochrome P450 family (CYPs) as drivers of TNBC. We examined by RNA sequencing the mTNBC and nadj parts of five women. We found more than a fivefold elevation in mTNBC of genes already known to be expressed in TNBC: BIRC5/survivin, Wnt-10A and -7B, matrix metalloproteinases (MMPs), chemokines, anterior gradient proteins, and lysophosphatidic acid receptor and the known basal characteristics of TNBC, sox10, ROPN1B, and Col9a3. There were two unexpected findings: 1) a strong induction of CYPs involved in activation of fatty acids (CYP4), and in inactivation of calcitriol (CYP24A1) and retinoic acid (CYP26A1); and 2) a marked down-regulation of FOS, FRA1, and JUN, known tethering partners of ERβ. ERβ is expressed in 20 to 30% of TNBCs and is being evaluated as a target for treating TNBC. We used ERβ+ TNBC patient-derived xenografts in mice and found that the ERβ agonist LY500703 had no effect on growth or proliferation. Expression of CYPs was confirmed by immunohistochemistry in formalin-fixed and paraffin-embedded (FFPE) TNBC. In TNBC cell lines, the CYP4Z1-catalyzed fatty acid metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) increased proliferation, while calcitriol decreased proliferation but only after inhibition of CYP24A1. We conclude that CYP-mediated pathways can be drivers of TNBC but that ERβ is unlikely to be a tumor suppressor because the absence of its main tethering partners renders ERβ functionless on genes involved in proliferation and inflammation.
Collapse
|
14
|
Yuan B, Yang J, Dubeau L, Hu Y, Li R. A Phosphotyrosine Switch in Estrogen Receptor β Is Required for Mouse Ovarian Function. Front Cell Dev Biol 2021; 9:649087. [PMID: 33898441 PMCID: PMC8063698 DOI: 10.3389/fcell.2021.649087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
The two homologous estrogen receptors ERα and ERβ exert distinct effects on their cognate tissues. Previous work from our laboratory identified an ERβ-specific phosphotyrosine residue that regulates ERβ transcriptional activity and antitumor function in breast cancer cells. To determine the physiological role of the ERβ phosphotyrosine residue in normal tissue development and function, we investigated a mutant mouse model (Y55F) whereby this particular tyrosine residue in endogenous mouse ERβ is mutated to phenylalanine. While grossly indistinguishable from their wild-type littermates, mutant female mice displayed reduced fertility, decreased ovarian follicular cell proliferation, and lower progesterone levels. Moreover, mutant ERβ from female mice during superovulation is defective in activating promoters of its target genes in ovarian tissues. Thus, our findings provide compelling genetic and molecular evidence for a role of isotype-specific ERβ phosphorylation in mouse ovarian development and function.
Collapse
Affiliation(s)
- Bin Yuan
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Jing Yang
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Louis Dubeau
- Department of Pathology, USC/Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA, United States
| | - Yanfen Hu
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Rong Li
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
15
|
Indukuri R, Jafferali MH, Song D, Damdimopoulos A, Hases L, Zhao C, Archer A, Williams C. Genome-wide estrogen receptor β chromatin binding in human colon cancer cells reveals its tumor suppressor activity. Int J Cancer 2021; 149:692-706. [PMID: 33754337 DOI: 10.1002/ijc.33573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer death in the western world. In women, menopausal hormone therapy has been shown to reduce CRC incidence by 20%. Studies demonstrate that estrogen activating estrogen receptor beta (ERβ) protects against CRC. ERβ is a nuclear receptor that regulates gene expression through interactions with the chromatin. This molecular mechanism is, however, not well characterized in colon. Here, we present for the first time, the cistrome of ERβ in different colon cancer cell lines. We use cell lines engineered to express ERβ, optimize and validate an ERβ antibody for chromatin-immunoprecipitation (ChIP), and perform ChIP-Seq. We identify key binding motifs, including ERE, AP-1, and TCF sites, and we determine enrichment of binding to cis-regulatory chromatin sites of genes involved in tumor development, cell migration, cell adhesion, apoptosis, and Wnt signaling pathways. We compare the corresponding cistromes of colon and breast cancer and find that they are conserved for about a third of genes, including GREB1, but that ERβ tethering to TCF and KLF family motifs is characteristic for colon. We exemplify upregulation of putative CRC tumor suppressor gene CST5 where ERβ in colon cells binds to cis-regulatory regions nearby (-351 bp) the transcriptional start site. Our work provides a foundation for understanding the mechanism of action of ERβ in CRC prevention.
Collapse
Affiliation(s)
- Rajitha Indukuri
- Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Mohammed Hakim Jafferali
- Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Dandan Song
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Anastasios Damdimopoulos
- Bioinformatics and Expression Core, Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Linnea Hases
- Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Amena Archer
- Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Cecilia Williams
- Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
16
|
Fels JA, Casalena GA, Manfredi G. Sex and oestrogen receptor β have modest effects on gene expression in the mouse brain posterior cortex. Endocrinol Diabetes Metab 2021; 4:e00191. [PMID: 33532622 PMCID: PMC7831211 DOI: 10.1002/edm2.191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/12/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Sex differences in brain cortical function affect cognition, behaviour and susceptibility to neural diseases, but the molecular basis of sexual dimorphism in cortical function is still largely unknown. Oestrogen and oestrogen receptors (ERs), specifically ERβ, the most abundant ER in the cortex, may play a role in determining sex differences in gene expression, which could underlie functional sex differences. However, further investigation is needed to address brain region specificity of the effects of sex and ERβ on gene expression. The goal of this study was to investigate sex differences in gene expression in the mouse posterior cortex, where sex differences in transcription have never been examined, and to determine how genetic ablation of ERβ affects transcription. Methods In this study, we performed unbiased transcriptomics on RNA from the posterior cortex of adult wild-type and ERβ knockout mice (n = 4/sex/genotype). We used unbiased clustering to analyse whole-transcriptome changes between the groups. We also performed differential expression analysis on the data using DESeq2 to identify specific changes in gene expression. Results We found only 27 significantly differentially expressed genes (DEGs) in wild-type (WT) males vs females, of which 17 were autosomal genes. Interestingly, in ERβKO males vs females all the autosomal DEGs were lost. Gene Ontology analysis of the subset of DEGs with sex differences only in the WT cortex revealed a significant enrichment of genes annotated with the function 'cation channel activity'. Moreover, within each sex we found only a few DEGs in ERβKO vs WT mice (8 and 5 in males and females, respectively). Conclusions Overall, our results suggest that in the adult mouse posterior cortex there are surprisingly few sex differences in gene expression, and those that exist are mainly related to cation channel activity. Additionally, they indicate that brain region-specific functional effects of ERβ may be largely post-transcriptional.
Collapse
Affiliation(s)
- Jasmine A. Fels
- Feil Family Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| | | | - Giovanni Manfredi
- Feil Family Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
17
|
Mal R, Magner A, David J, Datta J, Vallabhaneni M, Kassem M, Manouchehri J, Willingham N, Stover D, Vandeusen J, Sardesai S, Williams N, Wesolowski R, Lustberg M, Ganju RK, Ramaswamy B, Cherian MA. Estrogen Receptor Beta (ERβ): A Ligand Activated Tumor Suppressor. Front Oncol 2020; 10:587386. [PMID: 33194742 PMCID: PMC7645238 DOI: 10.3389/fonc.2020.587386] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) belong to a superfamily of nuclear receptors called steroid hormone receptors, which, upon binding ligand, dimerize and translocate to the nucleus where they activate or repress the transcription of a large number of genes, thus modulating critical physiologic processes. ERβ has multiple isoforms that show differing association with prognosis. Expression levels of the full length ERβ1 isoform are often lower in aggressive cancers as compared to normal tissue. High ERβ1 expression is associated with improved overall survival in women with breast cancer. The promise of ERβ activation, as a potential targeted therapy, is based on concurrent activation of multiple tumor suppressor pathways with few side effects compared to chemotherapy. Thus, ERβ is a nuclear receptor with broad-spectrum tumor suppressor activity, which could serve as a potential treatment target in a variety of human cancers including breast cancer. Further development of highly selective agonists that lack ERα agonist activity, will be necessary to fully harness the potential of ERβ.
Collapse
Affiliation(s)
- Rahul Mal
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Alexa Magner
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Joel David
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Jharna Datta
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Meghna Vallabhaneni
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Mahmoud Kassem
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jasmine Manouchehri
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Natalie Willingham
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Daniel Stover
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Jeffery Vandeusen
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Sagar Sardesai
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Nicole Williams
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Robert Wesolowski
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Maryam Lustberg
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Ramesh K Ganju
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Bhuvaneswari Ramaswamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| | - Mathew A Cherian
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States.,Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
18
|
Ventral prostate and mammary gland phenotype in mice with complete deletion of the ERβ gene. Proc Natl Acad Sci U S A 2020; 117:4902-4909. [PMID: 32075916 PMCID: PMC7060692 DOI: 10.1073/pnas.1920478117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Disagreements about the phenotype of estrogen receptor β (ERβ) knockout mouse, created by removing the DNA-binding domain of the ERβ gene or interruption of the gene with a neocassette (Oliver Smithies ERβ knockout mice [ERβOS-/-]), prompted us to create an ERβ knockout mouse by deleting the ERβ gene with the use of CRISPR/Cas9 technology. We confirmed that the ERβ gene was eliminated from the mouse genome and that no ERβ mRNA or protein was detectable in tissues of this mouse. Overall the phenotype of the ventral prostate (VP) and mammary gland (MG) in ERβcrispr-/- mice was similar to, but more severe than, that in the ERβOS-/-mice. In the VP of 6-mo-old ERβcrispr-/- mice there was epithelial hyperplasia, fibroplasia, inflammation, stromal overgrowth, and intraductal cancer-like lesions. This was accompanied by an increase in Ki67 and P63 and loss in DACH1 and PURα, two androgen receptor (AR) repressors. In the MG there was overexpression of estrogen receptor α and progesterone receptor, loss of collagen, increase in proliferation and expression of metalloproteases, and invasive epithelium. Surprisingly, by 18 mo of age, the number of hyperplastic foci was reduced, the ducts of the VP and MG became atrophic, and, in the VP, there was massive immune infiltration and massive desquamation of the luminal epithelial cells. These changes were coincident with reduced levels of androgens in males and estrogens in females. We conclude that ERβ is a tumor suppressor gene in the VP and MG where its loss increases the activity AR and ERα, respectively.
Collapse
|
19
|
Da Silva MHA, De Souza DB. Current evidence for the involvement of sex steroid receptors and sex hormones in benign prostatic hyperplasia. Res Rep Urol 2019; 11:1-8. [PMID: 30662879 PMCID: PMC6327899 DOI: 10.2147/rru.s155609] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is a pathology that affects 50% of men over 50 years of age and 90% of men develop BPH in their eighth decade of life. In 2018, more than 1 billion men will be affected by this disease worldwide. However, the progression of BPH is highly complex and has been debated and studied for approximately four decades. Recent studies indicate that BPH can originate from the alteration of different hormone synthesis pathways, and that it is also linked to the function of hormone receptors. There is a close relationship between the progression of BPH and sexual hormones, such as progesterone, testosterone, dihydrotestosterone, and estrogen. The focus of this study was to characterize the interactions of these hormones and investigate the direct or indirect role of each sex hormone receptor in the progression of BPH. Although several studies have described the effects of these hormones on BPH, no conclusions have been drawn regarding their role in disease progression. Here, we present a literature review on the sexual receptors possibly involved in the progression of BPH.
Collapse
|
20
|
Wang B, Li D, Rodriguez-Juarez R, Farfus A, Storozynsky Q, Malach M, Carpenter E, Filkowski J, Lykkesfeldt AE, Kovalchuk O. A suppressive role of guanine nucleotide-binding protein subunit beta-4 inhibited by DNA methylation in the growth of anti-estrogen resistant breast cancer cells. BMC Cancer 2018; 18:817. [PMID: 30103729 PMCID: PMC6090602 DOI: 10.1186/s12885-018-4711-0] [Citation(s) in RCA: 17] [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: 11/21/2017] [Accepted: 07/31/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Breast cancer is the most common malignancy in women worldwide. Although the endocrine therapy that targets estrogen receptor α (ERα) signaling has been well established as an effective adjuvant treatment for patients with ERα-positive breast cancers, long-term exposure may eventually lead to the development of acquired resistance to the anti-estrogen drugs, such as fulvestrant and tamoxifen. A better understanding of the mechanisms underlying antiestrogen resistance and identification of the key molecules involved may help in overcoming antiestrogen resistance in breast cancer. METHODS The whole-genome gene expression and DNA methylation profilings were performed using fulvestrant-resistant cell line 182R-6 and tamoxifen-resistant cell line TAMR-1 as a model system. In addition, qRT-PCR and Western blot analysis were performed to determine the levels of mRNA and protein molecules. MTT, apoptosis and cell cycle analyses were performed to examine the effect of either guanine nucleotide-binding protein beta-4 (GNB4) overexpression or knockdown on cell proliferation, apoptosis and cell cycle. RESULTS Among 9 candidate genes, GNB4 was identified and validated by qRT-PCR as a potential target silenced by DNA methylation via DNA methyltransferase 3B (DNMT3B). We generated stable 182R-6 and TAMR-1 cell lines that are constantly expressing GNB4 and determined the effect of the ectopic GNB4 on cell proliferation, cell cycle, and apoptosis of the antiestrogen-resistant cells in response to either fulvestrant or tamoxifen. Ectopic expression of GNB4 in two antiestrogen resistant cell lines significantly promoted cell growth and shortened cell cycle in the presence of either fulvestrant or tamoxifen. The ectopic GNB4 induced apoptosis in 182R-6 cells, whereas it inhibited apoptosis in TAMR-1 cells. Many regulators controlling cell cycle and apoptosis were aberrantly expressed in two resistant cell lines in response to the enforced GNB4 expression, which may contribute to GNB4-mediated biologic and/or pathologic processes. Furthermore, knockdown of GNB4 decreased growth of both antiestrogen resistant and sensitive breast cancer cells. CONCLUSION GNB4 is important for growth of breast cancer cells and a potential target for treatment.
Collapse
Affiliation(s)
- Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, People’s Republic of China
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, People’s Republic of China
| | | | - Allison Farfus
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
| | - Quinn Storozynsky
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
| | - Megan Malach
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
| | - Emily Carpenter
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
| | - Jody Filkowski
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
| | - Anne E. Lykkesfeldt
- Breast Cancer Group, Cell Death and Metabolism, Danish Cancer Society Research Center, Strandboulevarden, Copenhagen, Denmark
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB Canada
- Hepler Hall, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4 Canada
| |
Collapse
|
21
|
Estrogen enhances mismatch repair by induction of MLH1 expression via estrogen receptor-β. Oncotarget 2018; 8:38767-38779. [PMID: 28404976 PMCID: PMC5503570 DOI: 10.18632/oncotarget.16351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023] Open
Abstract
Epidemiological data demonstrated that hormone replace treatment has protective effect against colorectal cancer (CRC). Our previous studies showed that this effect may be associated with DNA mismatch repair. This study aims to investigate the mechanism of estrogen induction of MLH1, and whether colorectal tumor proliferation can be inhibited through induction of MLH1 by estrogen signal pathway. Human CRC cell lines were used to examine the regulation of MLH1 expression by over-expression and depletion of estrogen receptor-α (ERα) and estrogen receptor-β (ERβ), under the treatment with 17β-estradiol or β-Estradiol 6-(O-carboxy-methyl)oxime:BSA, followed by a real-time Q-PCR and Western blotting analysis. Luciferase reporter and chromatin immunoprecipitation assays were used to identify the estrogen response elements in the proximal promoter of MLH1 gene. Then, the influence of estrogen-induced MLH1 on CRC tumor growth were determined in vitro and in vivo. We found that mismatch repair ability and microsatellite stability of cells were enhanced by estrogen via induction of MLH1 expression, which was mediated by ERβ, through a transcriptional activation process. Furthermore, we identified that ERβ exerted an inhibitory effect on CRC tumor proliferation in vitro and in vivo, combined with 5-FU, through up-regulation of MLH1 expression. Finally, we concluded that estrogen enhances mismatch repair ability and tumor inhibition effect in vitro and in vivo, via induction of MLH1 expression mediated by ERβ.
Collapse
|
22
|
Mishra AK, Abrahamsson A, Dabrosin C. Fulvestrant inhibits growth of triple negative breast cancer and synergizes with tamoxifen in ERα positive breast cancer by up-regulation of ERβ. Oncotarget 2018; 7:56876-56888. [PMID: 27486755 PMCID: PMC5302959 DOI: 10.18632/oncotarget.10871] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/16/2016] [Indexed: 12/17/2022] Open
Abstract
The estrogen receptor-alpha (ERα) is used as a predictive marker for anti-estrogen therapy in breast cancer patients. In addition to aromatase inhibitors, ERα can be targeted at the receptor level using the receptor modulator tamoxifen or by the pure anti-estrogen fulvestrant. The role of the second ER, ER-beta (ERβ), as a therapeutic target or prognostic marker in breast cancer is still elusive. Hitherto, it is not known if ERα+/ERβ+ breast cancers would benefit from a treatment strategy combining tamoxifen and fulvestrant or if fulvestrant exert any therapeutic effects in ERα-/ERβ+ breast cancer. Here, we report that fulvestrant up-regulated ERβ in ERα+/ERβ+ breast cancer and in triple negative ERβ+ breast cancers (ERα-/ERβ+). In ERα+/ERβ+ breast cancer, a combination therapy of tamoxifen and fulvestrant significantly reduced tumor growth compared to either treatment alone both in vivo and in vitro. In ERα-/ERβ+ breast cancer fulvestrant had potent effects on cancer growth, in vivo as well as in vitro, and this effect was dependent on intrinsically expressed levels of ERβ. The role of ERβ was further confirmed in cells where ERβ was knocked-in or knocked-down. Inhibition of DNA methyltransferase (DNMT) increased the levels of ERβ and fulvestrant exerted similar potency on DNMT activity as the DNMT inhibitor decitabine. We conclude that fulvestrant may have therapeutic potential in additional groups of breast cancer patients; i) in ERα+/ERβ+ breast cancer where fulvestrant synergizes with tamoxifen and ii) in triple negative/ERβ+ breast cancer patients, a subgroup of breast cancer patients with poor prognosis.
Collapse
Affiliation(s)
- Ameet K Mishra
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Annelie Abrahamsson
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
23
|
Bado I, Nikolos F, Rajapaksa G, Gustafsson JÅ, Thomas C. ERβ decreases the invasiveness of triple-negative breast cancer cells by regulating mutant p53 oncogenic function. Oncotarget 2017; 7:13599-611. [PMID: 26871946 PMCID: PMC4924664 DOI: 10.18632/oncotarget.7300] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/29/2016] [Indexed: 01/06/2023] Open
Abstract
Most (80%) of the triple-negative breast cancers (TNBCs) express mutant p53 proteins that acquire oncogenic activities including promoting metastasis. We previously showed that wild-type ERβ (ERβ1) impedes epithelial to mesenchymal transition (EMT) and decreases the invasiveness of TNBC cells. In the present study we searched for signaling pathways that ERβ1 uses to inhibit EMT and invasion in TNBC cells. We show that ERβ1 binds to and opposes the transcriptional activity of mutant p53 at the promoters of genes that regulate metastasis. p63 that transcriptionally cooperates with mutant p53 also binds to ERβ1. Downregulation of p63 represses the epithelial phenotype of ERβ1-expressing cells and alters the expression of mutant p53 target genes. These results describe a novel mechanism through which ERβ1 can disturb oncogenic signals to inhibit aggressiveness in TNBCs.
Collapse
Affiliation(s)
- Igor Bado
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas 77204, USA
| | - Fotis Nikolos
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas 77204, USA
| | - Gayani Rajapaksa
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas 77204, USA
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas 77204, USA
| | - Christoforos Thomas
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas 77204, USA
| |
Collapse
|
24
|
Role of estrogen receptor beta in neural differentiation of mouse embryonic stem cells. Proc Natl Acad Sci U S A 2017; 114:E10428-E10437. [PMID: 29133394 DOI: 10.1073/pnas.1714094114] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The ability to propagate mature cells and tissue from pluripotent stem cells offers enormous promise for treating many diseases, including neurodegenerative diseases. Before such cells can be used successfully in neurodegenerative diseases without causing unwanted cell growth and migration, genes regulating growth and migration of neural stem cells need to be well characterized. Estrogen receptor beta (ERβ) is essential for migration of neurons and glial cells in the developing mouse brain. To examine whether ERβ influences differentiation of mouse embryonic stem cells (mESC) into neural lineages, we compared control and ERβ knockout (BERKO) mESCs at defined stages of neural development and examined the effects of an ERβ-selective ligand (LY3201) with a combination of global and targeted gene-expression profiling and the expression of key pluripotency markers. We found that ERβ was induced in embryoid bodies (EBs) and neural precursor cells (NPCs) during development. Proliferation was higher in BERKO NPCs and was inhibited by LY3201. Neurogenesis was reduced in BERKO ES cells, and oligodendrogliogenesis was enhanced. BERKO EBs expressed higher levels of key ectodermal and neural progenitor markers and lower levels of markers for mesoderm and endoderm lineages. ERβ-regulated factors are involved in cell adhesion, axon guidance, and signaling of Notch and GABA receptor pathways, as well as factors important for the differentiation of neuronal precursors into dopaminergic neurons (Engrailed 1) and for the oligodendrocyte fate acquisition (Olig2). Our data suggest that ERβ is an important component for differentiation into midbrain neurons as well as for preventing precocious oligodendrogliogenesis.
Collapse
|
25
|
Miller MM, McMullen PD, Andersen ME, Clewell RA. Multiple receptors shape the estrogen response pathway and are critical considerations for the future of in vitro-based risk assessment efforts. Crit Rev Toxicol 2017; 47:564-580. [DOI: 10.1080/10408444.2017.1289150] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
26
|
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
|
27
|
Guo Y, Wang Q, Li G, He M, Tang H, Zhang H, Yang X, Liu X, Lin H. Molecular mechanism of feedback regulation of 17β-estradiol on two kiss
genes in the protogynous orange-spotted grouper (Epinephelus coioides
). Mol Reprod Dev 2017; 84:495-507. [DOI: 10.1002/mrd.22800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/20/2017] [Indexed: 02/05/2023]
Affiliation(s)
- Yin Guo
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences; Sun Yat-Sen University; Guangzhou China
| | - Qingqing Wang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences; Sun Yat-Sen University; Guangzhou China
| | - Gaofei Li
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences; Sun Yat-Sen University; Guangzhou China
| | - Meng He
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences; Sun Yat-Sen University; Guangzhou China
| | - Haipei Tang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences; Sun Yat-Sen University; Guangzhou China
| | - Haifa Zhang
- Marine Fisheries Development Center of Guangdong Province; Huizhou China
| | - Xiaoli Yang
- Key Laboratory of Aquatic Animal Breeding and Culturing for South China Sea; Ministry of Agriculture; Guangzhou China
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences; Sun Yat-Sen University; Guangzhou China
- South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center; Guangzhou China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences; Sun Yat-Sen University; Guangzhou China
- South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center; Guangzhou China
| |
Collapse
|
28
|
Understanding the dynamics of Toll-like Receptor 5 response to flagellin and its regulation by estradiol. Sci Rep 2017; 7:40981. [PMID: 28112187 PMCID: PMC5256028 DOI: 10.1038/srep40981] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/13/2016] [Indexed: 12/24/2022] Open
Abstract
Toll-like receptors (TLRs) are major players of the innate immune system. Once activated, they trigger a signalling cascade that leads to NF-κB translocation from the cytoplasm to the nucleus. Single cell analysis shows that NF-κB signalling dynamics are a critical determinant of transcriptional regulation. Moreover, the outcome of innate immune response is also affected by the cross-talk between TLRs and estrogen signalling. Here, we characterized the dynamics of TLR5 signalling, responsible for the recognition of flagellated bacteria, and those changes induced by estradiol in its signalling at the single cell level. TLR5 activation in MCF7 cells induced a single and sustained NF-κB translocation into the nucleus that resulted in high NF-κB transcription activity. The overall magnitude of NF-κB transcription activity was not influenced by the duration of the stimulus. No significant changes are observed in the dynamics of NF-κB translocation to the nucleus when MCF7 cells are incubated with estradiol. However, estradiol significantly decreased NF-κB transcriptional activity while increasing TLR5-mediated AP-1 transcription. The effect of estradiol on transcriptional activity was dependent on the estrogen receptor activated. This fine tuning seems to occur mainly in the nucleus at the transcription level rather than affecting the translocation of the NF-κB transcription factor.
Collapse
|
29
|
Miao YF, Su W, Dai YB, Wu WF, Huang B, Barros RPA, Nguyen H, Maneix L, Guan YF, Warner M, Gustafsson JÅ. An ERβ agonist induces browning of subcutaneous abdominal fat pad in obese female mice. Sci Rep 2016; 6:38579. [PMID: 27922125 PMCID: PMC5138613 DOI: 10.1038/srep38579] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/09/2016] [Indexed: 02/07/2023] Open
Abstract
Estrogen, via estrogen receptor alpha (ERα), exerts several beneficial effects on metabolism and energy homeostasis by controlling size, enzymatic activity and hormonal content of adipose tissue. The actions of estrogen on sympathetic ganglia, which are key players in the browning process, are less well known. In the present study we show that ERβ influences browning of subcutaneous adipose tissue (SAT) via its actions both on sympathetic ganglia and on the SAT itself. A 3-day-treatment with a selective ERβ agonist, LY3201, induced browning of SAT in 1-year-old obese WT and ERα−/− female mice. Browning was associated with increased expression of ERβ in the nuclei of neurons in the sympathetic ganglia, increase in tyrosine hydroxylase in both nerve terminals in the SAT and sympathetic ganglia neurons and an increase of β3-adrenoceptor in the SAT. LY3201 had no effect on browning in young female or male mice. In the case of young females browning was already maximal while in males there was very little expression of ERβ in the SAT and very little expression of the β3-adrenoceptor. The increase in both sympathetic tone and responsiveness of adipocytes to catecholamines reveals a novel role for ERβ in controlling browning of adipose tissue.
Collapse
Affiliation(s)
- Yi-Fei Miao
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Wen Su
- Center for Nephrology and Urology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Yu-Bing Dai
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Wan-Fu Wu
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Bo Huang
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Rodrigo P A Barros
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Hao Nguyen
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Laure Maneix
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - You-Fei Guan
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.,Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Margaret Warner
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.,Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet, Novum, 14186 Stockholm, Sweden
| |
Collapse
|
30
|
Pendse SN, Maertens A, Rosenberg M, Roy D, Fasani RA, Vantangoli MM, Madnick SJ, Boekelheide K, Fornace AJ, Odwin SA, Yager JD, Hartung T, Andersen ME, McMullen PD. Information-dependent enrichment analysis reveals time-dependent transcriptional regulation of the estrogen pathway of toxicity. Arch Toxicol 2016; 91:1749-1762. [PMID: 27592001 PMCID: PMC5364265 DOI: 10.1007/s00204-016-1824-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/22/2016] [Indexed: 02/06/2023]
Abstract
The twenty-first century vision for toxicology involves a transition away from high-dose animal studies to in vitro and computational models (NRC in Toxicity testing in the 21st century: a vision and a strategy, The National Academies Press, Washington, DC, 2007). This transition requires mapping pathways of toxicity by understanding how in vitro systems respond to chemical perturbation. Uncovering transcription factors/signaling networks responsible for gene expression patterns is essential for defining pathways of toxicity, and ultimately, for determining the chemical modes of action through which a toxicant acts. Traditionally, transcription factor identification is achieved via chromatin immunoprecipitation studies and summarized by calculating which transcription factors are statistically associated with up- and downregulated genes. These lists are commonly determined via statistical or fold-change cutoffs, a procedure that is sensitive to statistical power and may not be as useful for determining transcription factor associations. To move away from an arbitrary statistical or fold-change-based cutoff, we developed, in the context of the Mapping the Human Toxome project, an enrichment paradigm called information-dependent enrichment analysis (IDEA) to guide identification of the transcription factor network. We used a test case of activation in MCF-7 cells by 17β estradiol (E2). Using this new approach, we established a time course for transcriptional and functional responses to E2. ERα and ERβ were associated with short-term transcriptional changes in response to E2. Sustained exposure led to recruitment of additional transcription factors and alteration of cell cycle machinery. TFAP2C and SOX2 were the transcription factors most highly correlated with dose. E2F7, E2F1, and Foxm1, which are involved in cell proliferation, were enriched only at 24 h. IDEA should be useful for identifying candidate pathways of toxicity. IDEA outperforms gene set enrichment analysis (GSEA) and provides similar results to weighted gene correlation network analysis, a platform that helps to identify genes not annotated to pathways.
Collapse
Affiliation(s)
- Salil N Pendse
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA.,ScitoVation, LLC, 6 Davis Drive, PO Box 110566, Research Triangle Park, NC, 27709, USA
| | - Alexandra Maertens
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | - Samantha J Madnick
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Albert J Fornace
- Department of Biochemistry and Molecular and Cellular Biology, and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Shelly-Ann Odwin
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - James D Yager
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.,Center for Alternatives to Animal Testing-Europe, University of Konstanz, Constance, Germany
| | - Melvin E Andersen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA.,ScitoVation, LLC, 6 Davis Drive, PO Box 110566, Research Triangle Park, NC, 27709, USA
| | - Patrick D McMullen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA. .,ScitoVation, LLC, 6 Davis Drive, PO Box 110566, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
31
|
Yuan B, Cheng L, Gupta K, Chiang HC, Gupta HB, Sareddy GR, Wang D, Lathrop K, Elledge R, Wang P, McHardy S, Vadlamudi R, Curiel TJ, Hu Y, Ye Q, Li R. Tyrosine phosphorylation regulates ERβ ubiquitination, protein turnover, and inhibition of breast cancer. Oncotarget 2016; 7:42585-42597. [PMID: 27323858 PMCID: PMC5173158 DOI: 10.18632/oncotarget.10018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 05/23/2016] [Indexed: 12/31/2022] Open
Abstract
Unlike estrogen receptor α (ERα) that predominantly promotes hormone-dependent breast tumor growth, ERβ exhibits antitumor effects in a variety of cancer types. We recently identified a phosphotyrosine residue in ERβ, but not ERα, that dictates ERβ transcriptional activity and antitumor function. We show here that this ER isotype-specific phosphotyrosine switch is important for regulating ERβ activity in cell proliferation, migration, and invasion. At the mechanistic level, phosphorylated ERβ, which recruits transcriptional coactivator p300, is in turn targeted by p300 for ubiquitination and proteasome-dependent protein turnover. Furthermore, ERβ-specific agonists such as S-equol enhance ERβ phosphorylation, suggesting a crosstalk between ligand- and posttranslational modification-dependent ERβ activation. Inhibition of xenograft tumor growth by S-equol is associated with reduced tumor Ki-67 expression and elevated ERβ tyrosine phosphorylation. Taken together, our data support the notion that phosphotyrosine-dependent ERβ signaling is an attractive target for anticancer treatment.
Collapse
Affiliation(s)
- Bin Yuan
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, China
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Long Cheng
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Kshama Gupta
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Huai-Chin Chiang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Harshita B. Gupta
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Degeng Wang
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kate Lathrop
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Richard Elledge
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Pei Wang
- Department of Cellular and Structural Biology Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stanton McHardy
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, USA
| | - Ratna Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tyler J. Curiel
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yanfen Hu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, China
| | - Rong Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
32
|
Sareddy GR, Li X, Liu J, Viswanadhapalli S, Garcia L, Gruslova A, Cavazos D, Garcia M, Strom AM, Gustafsson JA, Tekmal RR, Brenner A, Vadlamudi RK. Selective Estrogen Receptor β Agonist LY500307 as a Novel Therapeutic Agent for Glioblastoma. Sci Rep 2016; 6:24185. [PMID: 27126081 PMCID: PMC4850367 DOI: 10.1038/srep24185] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/21/2016] [Indexed: 02/07/2023] Open
Abstract
Glioblastomas (GBM), deadly brain tumors, have greater incidence in males than females. Epidemiological evidence supports a tumor suppressive role of estrogen; however, estrogen as a potential therapy for GBM is limited due to safety concerns. Since GBM express ERβ, a second receptor for estrogen, targeting ERβ with a selective agonist may be a potential novel GBM therapy. In the present study, we examined the therapeutic effect of the selective synthetic ERβ agonist LY500307 using in vitro and in vivo GBM models. Treatment with LY500307 significantly reduced the proliferation of GBM cells with no activity on normal astrocytes in vitro. ERβ agonists promoted apoptosis of GBM cells, and mechanistic studies using RNA sequencing revealed that LY500307 modulated several pathways related to apoptosis, cell cycle, and DNA damage response. Further, LY500307 sensitized GBM cells to several FDA-approved chemotherapeutic drugs including cisplatin, lomustine and temozolomide. LY500307 treatment significantly reduced the in vivo tumor growth and promoted apoptosis of GBM tumors in an orthotopic model and improved the overall survival of tumor-bearing mice in the GL26 syngeneic glioma model. Our results demonstrate that LY500307 has potential as a therapeutic agent for GBM.
Collapse
Affiliation(s)
- Gangadhara R Sareddy
- The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Xiaonan Li
- The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Jinyou Liu
- The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Suryavathi Viswanadhapalli
- The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Lauren Garcia
- The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Aleksandra Gruslova
- Cancer Therapy &Research Center, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - David Cavazos
- Cancer Therapy &Research Center, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Mike Garcia
- Cancer Therapy &Research Center, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | | | | | - Rajeshwar Rao Tekmal
- The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Andrew Brenner
- Cancer Therapy &Research Center, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA.,Hematology &Oncology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| | - Ratna K Vadlamudi
- The Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA.,Cancer Therapy &Research Center, University of Texas Health Science Center at San Antonio, San Antonio TX 78229, USA
| |
Collapse
|
33
|
Nakajima Y, Osakabe A, Waku T, Suzuki T, Akaogi K, Fujimura T, Homma Y, Inoue S, Yanagisawa J. Estrogen Exhibits a Biphasic Effect on Prostate Tumor Growth through the Estrogen Receptor β-KLF5 Pathway. Mol Cell Biol 2016; 36:144-56. [PMID: 26483416 PMCID: PMC4702593 DOI: 10.1128/mcb.00625-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 07/14/2015] [Accepted: 10/09/2015] [Indexed: 11/20/2022] Open
Abstract
Estrogens are effective in the treatment of prostate cancer; however, the effects of estrogens on prostate cancer are enigmatic. In this study, we demonstrated that estrogen (17β-estradiol [E2]) has biphasic effects on prostate tumor growth. A lower dose of E2 increased tumor growth in mouse xenograft models using DU145 and PC-3 human prostate cancer cells, whereas a higher dose significantly decreased tumor growth. We found that anchorage-independent apoptosis in these cells was inhibited by E2 treatment. Similarly, in vivo angiogenesis was suppressed by E2. Interestingly, these effects of E2 were abolished by knockdown of either estrogen receptor β (ERβ) or Krüppel-like zinc finger transcription factor 5 (KLF5). Ιn addition, E2 suppressed KLF5-mediated transcription through ERβ, which inhibits proapoptotic FOXO1 and proangiogenic PDGFA expression. Furthermore, we revealed that a nonagonistic ER ligand GS-1405 inhibited FOXO1 and PDGFA expression through the ERβ-KLF5 pathway and regulated prostate tumor growth without ERβ transactivation. Therefore, these results suggest that E2 biphasically modulates prostate tumor formation by regulating KLF5-dependent transcription through ERβ and provide a new strategy for designing ER modulators, which will be able to regulate prostate cancer progression with minimal adverse effects due to ER transactivation.
Collapse
Affiliation(s)
- Yuka Nakajima
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Asami Osakabe
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tsuyoshi Waku
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Kensuke Akaogi
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tetsuya Fujimura
- Department of Urology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yukio Homma
- Department of Urology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Satoshi Inoue
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan Department of Anti-Aging Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Junn Yanagisawa
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
34
|
Klinge CM. miRNAs regulated by estrogens, tamoxifen, and endocrine disruptors and their downstream gene targets. Mol Cell Endocrinol 2015; 418 Pt 3:273-97. [PMID: 25659536 PMCID: PMC4523495 DOI: 10.1016/j.mce.2015.01.035] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are short (22 nucleotides), single-stranded, non-coding RNAs that form complimentary base-pairs with the 3' untranslated region of target mRNAs within the RNA-induced silencing complex (RISC) and block translation and/or stimulate mRNA transcript degradation. The non-coding miRBase (release 21, June 2014) reports that human genome contains ∼ 2588 mature miRNAs which regulate ∼ 60% of human protein-coding mRNAs. Dysregulation of miRNA expression has been implicated in estrogen-related diseases including breast cancer and endometrial cancer. The mechanism for estrogen regulation of miRNA expression and the role of estrogen-regulated miRNAs in normal homeostasis, reproduction, lactation, and in cancer is an area of great research and clinical interest. Estrogens regulate miRNA transcription through estrogen receptors α and β in a tissue-specific and cell-dependent manner. This review focuses primarily on the regulation of miRNA expression by ligand-activated ERs and their bona fide gene targets and includes miRNA regulation by tamoxifen and endocrine disrupting chemicals (EDCs) in breast cancer and cell lines.
Collapse
Affiliation(s)
- Carolyn M Klinge
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|
35
|
Pinceti E, Shults CL, Rao YS, Mott NN, Pak TR. Phosphorylation Alters Oestrogen Receptor β-Mediated Transcription in Neurones. J Neuroendocrinol 2015; 27:861-71. [PMID: 26440063 PMCID: PMC4661120 DOI: 10.1111/jne.12326] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 09/21/2015] [Accepted: 09/29/2015] [Indexed: 02/02/2023]
Abstract
Nuclear steroid hormone receptors are ubiquitously expressed transcription factors whose activity can be altered by post-translational modifications, such as phosphorylation. The consequences of post-translational modifications have been described for several members of the nuclear steroid hormone receptor superfamily; however, little is known about the effects of oestrogen receptor (ER)β phosphorylation in the brain. Moreover, to our knowledge, the presence of phosphorylated ERβ has not been detected in the brain of any species to date. Oestrogen receptor β is highly expressed in several regions of the brain and in vitro studies have demonstrated that it can be phosphorylated at two serine residues (S87 and S105) in the N-terminal AF-1 region. The present study aimed to determine whether phosphorylated ERβ is detectable in the hippocampus of aged female rats, as well as the functional consequences of ERβ S87 and S105 phosphorylation on transcriptional activity in neuronal cells. First, we used a novel PhosTag(™) approach to detect phosphorylated forms of ERβ in the dorsal hippocampus of aged female rats. The data obtained demonstrated abundant forms of phosphorylated ERβ in the dorsal hippocampus, suggesting that this post-translational modification might be an important regulator of ERβ function. To assess the functional consequences of ERβ phosphorylation in neuronal cells, we created phospho-mimetic (S87E, S105E) and phospho-null (S87A, S105A) ERβ receptors that were transiently transfected in a hippocampal-derived cell line. Collectively, our results showed that phosphorylation of S87 and S105 altered both ligand-independent and ligand-dependent ERβ transcriptional regulation. Overall, these data demonstrate that phosphorylated forms of ERβ are present in the brain of aged female rats and that phosphorylation of ERβ could differentially alter ERβ-mediated gene expression.
Collapse
Affiliation(s)
- E Pinceti
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - C L Shults
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Y S Rao
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - N N Mott
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - T R Pak
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
36
|
Abstract
Estrogen receptors alpha (ERα) and beta (ERβ) are transcription factors that are involved in the regulation of many complex physiological processes in humans. Abnormal ER signaling leads to development of a variety of diseases, such as cancer, metabolic and cardiovascular disease, neurodegeneration, inflammation, and osteoporosis. This review provides an overview and update on ERα and ERβ in health and disease with focus on their role in cancer and metabolic disease and in the context of recent years' success in providing genome wide data on ER function. Furthermore, potential clinical applications and challenges are also discussed.
Collapse
Affiliation(s)
- Min Jia
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, S-141 83 Huddinge, Sweden.
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, S-141 83 Huddinge, Sweden; SciLifeLab, Department of Biosciences and Nutrition, Karolinska Institutet, S-171 21 Solna, Sweden.
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, S-141 83 Huddinge, Sweden; Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, 3605 Cullen Blvd. Science and Engineering Research Center Bldg. 545, Houston, TX 77204-5056, United States.
| |
Collapse
|
37
|
Yun H, Xie J, Olumi AF, Ghosh R, Kumar AP. Activation of AKR1C1/ERβ induces apoptosis by downregulation of c-FLIP in prostate cancer cells: A prospective therapeutic opportunity. Oncotarget 2015; 6:11600-13. [PMID: 25816367 PMCID: PMC4484479 DOI: 10.18632/oncotarget.3417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/19/2015] [Indexed: 12/29/2022] Open
Abstract
We provide first-time evidence for ERβ-mediated transcriptional upregulation of c-FLIP as an underlying mechanism in the development of castrate-resistant cancer. While androgens inhibit apoptosis partly through transcriptional upregulation of the anti-apoptotic protein, c-FLIP in androgen-responsive cells, they downregulate c-FLIP in androgen-independent cells. We found that although Sp1 and p65 trans-activate c-FLIP, the combination of Sp1 and p65 has differential effects in a cellular context-dependent manner. We show that activation of the androgen metabolism enzyme, aldo-keto reductase, AKR1C1, relieves androgen independence through activation of 3β-Adiol-mediated upregulation of ERβ. ERβ competes with Sp1 and Sp3 to transcriptionally downregulate c-FLIP in the absence of consensus estrogen-response element in androgen-independent cells. Forced expression of AR in androgen-independent cells show that ERβ-mediated growth inhibition occurs under conditions of androgen independence. Reactivation of ERβ with the estrogenic metabolite, 2-methoxyestradiol, decreased enrichment ratio of Sp1/Sp3 at the c-FLIP promoter with concomitant effects on cell growth and death. Expression of Sp1 and c-FLIP are elevated while AKR1C1, ERβ and Sp3 are significantly low in human prostate tumor samples. ERβ is epigenetically silenced in prostate cancer patients, therefore our results suggest that combination of ERβ agonists with ADT would benefit men stratified on the basis of high estrogen levels.
Collapse
Affiliation(s)
- Huiyoung Yun
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Jianping Xie
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Urology, Shanxi Dayi Hospital, Shanxi Academy of Medical Science, Taiyuan, P.R., China
| | - Aria F. Olumi
- Department of Urology, Massachusetts General Hospital Harvard Medical School, Boston, MA, USA
| | - Rita Ghosh
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Molecular Medicine, The University of Texas Health Science Center, San Antonio, TX, USA
- Cancer Therapy and Research Center, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Addanki P. Kumar
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Molecular Medicine, The University of Texas Health Science Center, San Antonio, TX, USA
- Cancer Therapy and Research Center, The University of Texas Health Science Center, San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
| |
Collapse
|
38
|
Estrogen receptor β exon 3-deleted mouse: The importance of non-ERE pathways in ERβ signaling. Proc Natl Acad Sci U S A 2015; 112:5135-40. [PMID: 25848008 DOI: 10.1073/pnas.1504944112] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In 1998, an estrogen receptor β (ERβ) knockout (KO) mouse was created by interrupting the gene at the DNA binding domain (DBD) with a neocassette. The mutant females were subfertile and there were abnormalities in the brain, prostate, lung, colon, and immune system. In 2008, another ERβ mutant mouse was generated by deleting ERβ exon 3 which encodes the first zinc finger in the DBD. The female mice of this strain were unable to ovulate but were otherwise normal. The differences in the phenotypes of the two KO strains, have led to questions about the physiological function of ERβ. In the present study, we created an ERβ exon 3-deleted mouse (ERβ-Δex3) and confirmed that the only observable defect was anovulation. Despite the two in-frame stop codons introduced by splicing between exons 2 and 4, an ERβ protein was expressed in nuclei of prostate epithelial cells. Using two different anti-ERβ antibodies, we showed that an in-frame ligand binding domain and C terminus were present in the ERβ-Δex3 protein. Moreover, with nuclear extracts from ERβ-Δex3 prostates, there was an ERβ-dependent retardation of migration of activator protein-1 response elements in EMSA. Unlike the original knockout mouse, expression of Ki67, androgen receptor, and Dachshund-1 in prostate epithelium was not altered in the ERβ-Δex3 mouse. We conclude that very little of ERβ transcriptional activity depends on binding to classical estrogen response elements (EREs).
Collapse
|
39
|
Abstract
Prostate cancer is the commonest, non-cutaneous cancer in men. At present, there is no cure for the advanced, castration-resistant form of the disease. Estrogen has been shown to be important in prostate carcinogenesis, with evidence resulting from epidemiological, cancer cell line, human tissue and animal studies. The prostate expresses both estrogen receptor alpha (ERA) and estrogen receptor beta (ERB). Most evidence suggests that ERA mediates the harmful effects of estrogen in the prostate, whereas ERB is tumour suppressive, but trials of ERB-selective agents have not translated into improved clinical outcomes. The role of ERB in the prostate remains unclear and there is increasing evidence that isoforms of ERB may be oncogenic. Detailed study of ERB and ERB isoforms in the prostate is required to establish their cell-specific roles, in order to determine if therapies can be directed towards ERB-dependent pathways. In this review, we summarise evidence on the role of ERB in prostate cancer and highlight areas for future research.
Collapse
Affiliation(s)
- Adam W Nelson
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - Wayne D Tilley
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - David E Neal
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - Jason S Carroll
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| |
Collapse
|
40
|
Monsivais D, Dyson MT, Yin P, Coon JS, Navarro A, Feng G, Malpani SS, Ono M, Ercan CM, Wei JJ, Pavone ME, Su E, Bulun SE. ERβ- and prostaglandin E2-regulated pathways integrate cell proliferation via Ras-like and estrogen-regulated growth inhibitor in endometriosis. Mol Endocrinol 2014; 28:1304-15. [PMID: 24992181 DOI: 10.1210/me.2013-1421] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In endometriosis, stromal and epithelial cells from the endometrium form extrauterine lesions and persist in response to estrogen (E2) and prostaglandin E2 (PGE2). Stromal cells produce excessive quantities of estrogen and PGE2 in a feed-forward manner. However, it is unknown how estrogen stimulates cell proliferation and survival for the establishment and persistence of disease. Previous studies suggest that estrogen receptor-β (ERβ) is strikingly overexpressed in endometriotic stromal cells. Thus, we integrated genome-wide ERβ binding data from previously published studies in breast cells and gene expression profiles in human endometriosis and endometrial tissues (total sample number = 81) and identified Ras-like, estrogen-regulated, growth inhibitor (RERG) as an ERβ target. Estradiol potently induced RERG mRNA and protein levels in primary endometriotic stromal cells. Chromatin immunoprecipitation demonstrated E2-induced enrichment of ERβ at the RERG promoter region. PGE2 via protein kinase A phosphorylated RERG and enhanced the nuclear translocation of RERG. RERG induced the proliferation of primary endometriotic cells. Overall, we demonstrated that E2/ERβ and PGE2 integrate at RERG, leading to increased endometriotic cell proliferation and represents a novel candidate for therapeutic intervention.
Collapse
Affiliation(s)
- D Monsivais
- Division of Reproductive Biology Research (D.M., M.T.D., P.Y., J.S.C., A.N., S.S.M., M.O., C.M.E., M.E.P., E.S., S.E.B.), Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Northwestern University Biomedical Informatics Center (part of the Northwestern CTSA) and The Robert H. Lurie Comprehensive Cancer Center (G.F.), and Department of Pathology (J.J.W.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bondesson M, Hao R, Lin CY, Williams C, Gustafsson JÅ. Estrogen receptor signaling during vertebrate development. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:142-51. [PMID: 24954179 DOI: 10.1016/j.bbagrm.2014.06.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 01/03/2023]
Abstract
Estrogen receptors are expressed and their cognate ligands produced in all vertebrates, indicative of important and conserved functions. Through evolution estrogen has been involved in controlling reproduction, affecting both the development of reproductive organs and reproductive behavior. This review broadly describes the synthesis of estrogens and the expression patterns of aromatase and the estrogen receptors, in relation to estrogen functions in the developing fetus and child. We focus on the role of estrogens for the development of reproductive tissues, as well as non-reproductive effects on the developing brain. We collate data from human, rodent, bird and fish studies and highlight common and species-specific effects of estrogen signaling on fetal development. Morphological malformations originating from perturbed estrogen signaling in estrogen receptor and aromatase knockout mice are discussed, as well as the clinical manifestations of rare estrogen receptor alpha and aromatase gene mutations in humans. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Maria Bondesson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA.
| | - Ruixin Hao
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA; DuPont Haskell Global Centers for Health and Environmental Sciences, Newark, DE, USA
| | - Chin-Yo Lin
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA
| | - Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA; Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| |
Collapse
|
42
|
Zhao C, Qiao Y, Jonsson P, Wang J, Xu L, Rouhi P, Sinha I, Cao Y, Williams C, Dahlman-Wright K. Genome-wide profiling of AP-1-regulated transcription provides insights into the invasiveness of triple-negative breast cancer. Cancer Res 2014; 74:3983-94. [PMID: 24830720 DOI: 10.1158/0008-5472.can-13-3396] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive clinical subtype accounting for up to 20% of all breast cancers, but its malignant determinants remain largely undefined. Here, we show that in TNBC the overexpression of Fra-1, a component of the transcription factor AP-1, offers prognostic potential. Fra-1 depletion or its heterodimeric partner c-Jun inhibits the proliferative and invasive phenotypes of TNBC cells in vitro. Similarly, RNAi-mediated attenuation of Fra-1 or c-Jun reduced cellular invasion in vivo in a zebrafish tumor xenograft model. Exploring the AP-1 cistrome and the AP-1-regulated transcriptome, we obtained insights into the transcriptional regulatory networks of AP-1 in TNBC cells. Among the direct targets identified for Fra-1/c-Jun involved in proliferation, adhesion, and cell-cell contact, we found that AP-1 repressed the expression of E-cadherin by transcriptional upregulation of ZEB2 to stimulate cell invasion. Overall, this work illuminates the pathways through which TNBC cells acquire invasive and proliferative properties.
Collapse
Affiliation(s)
- Chunyan Zhao
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge;
| | - Yichun Qiao
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge
| | - Philip Jonsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas; and
| | - Jian Wang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm
| | - Li Xu
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge
| | - Pegah Rouhi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm
| | - Indranil Sinha
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm; Department of Medicine and Health Sciences, Linköping University, Linköping; Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
| | - Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas; and
| | - Karin Dahlman-Wright
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge; Science for Life Laboratory, Karolinska Institute, Solna, Sweden;
| |
Collapse
|
43
|
Jonsson P, Katchy A, Williams C. Support of a bi-faceted role of estrogen receptor β (ERβ) in ERα-positive breast cancer cells. Endocr Relat Cancer 2014; 21:143-60. [PMID: 24192230 PMCID: PMC3946733 DOI: 10.1530/erc-13-0444] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The expression of estrogen receptor α (ERα) in breast cancer identifies patients most likely to respond to endocrine treatment. The second ER, ERβ, is also expressed in breast tumors, but its function and therapeutic potential need further study. Although in vitro studies have established that ERβ opposes transcriptional and proliferative functions of ERα, several clinical studies report its correlation with proliferative markers and poorer prognosis. The data demonstrate that ERβ opposes ERα are primarily based on transient expression of ERβ. Here, we explored the functions of constitutively expressed ERβ in ERα-positive breast cancer lines MCF7 and T47D. We found that ERβ, under these conditions heterodimerized with ERα in the presence and absence of 17β-estradiol, and induced genome-wide transcriptional changes. Widespread anti-ERα signaling was, however, not observed and ERβ was not antiproliferative. Tamoxifen antagonized proliferation and ER-mediated gene regulation both in the presence and absence of ERβ. In conclusion, ERβ's role in cells adapted to its expression appears to differ from its role in cells with transient expression. Our study is important because it provides a deeper understanding of ERβ's role in breast tumors that coexpress both receptors and supports an emerging bi-faceted role of ERβ.
Collapse
Affiliation(s)
| | | | - Cecilia Williams
- To whom correspondence should be addressed:, Postal address: Center for Nuclear Receptors and Cell Signaling, 3605 Cullen Blvd., SERC Bldg. 545, Houston, TX 77204-5056,
| |
Collapse
|
44
|
Dey P, Barros RPA, Warner M, Ström A, Gustafsson JÅ. Insight into the mechanisms of action of estrogen receptor β in the breast, prostate, colon, and CNS. J Mol Endocrinol 2013; 51:T61-74. [PMID: 24031087 DOI: 10.1530/jme-13-0150] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Estrogen and its receptors (ERs) influence many biological processes in physiology and pathology in men and women. ERs are involved in the etiology and/or progression of cancers of the prostate, breast, uterus, ovary, colon, lung, stomach, and malignancies of the immune system. In estrogen-sensitive malignancies, ERβ usually is a tumor suppressor and ERα is an oncogene. ERβ regulates genes in several key pathways including tumor suppression (p53, PTEN); metabolism (PI3K); survival (Akt); proliferation pathways (p45(Skp2), cMyc, and cyclin E); cell-cycle arresting factors (p21(WAF1), cyclin-dependent kinase inhibitor 1 (CDKN1A)), p27(Kip1), and cyclin-dependent kinases (CDKs); protection from reactive oxygen species, glutathione peroxidase. Because they are activated by small molecules, ERs are excellent targets for pharmaceuticals. ERα antagonists have been used for many years in the treatment of breast cancer and more recently pharmaceutical companies have produced agonists which are very selective for ERα or ERβ. ERβ agonists are being considered for preventing progression of cancer, treatment of anxiety and depression, as anti-inflammatory agents and as agents, which prevent or reduce the severity of neurodegenerative diseases.
Collapse
Affiliation(s)
- Prasenjit Dey
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Blvd, Science and Engineering Research Center Bldg 545, Houston, Texas 77204-5056, USA Department of Biosciences and Nutrition, Karolinska Institutet, Novum, S-141 57 Huddinge, Sweden
| | | | | | | | | |
Collapse
|
45
|
Williams C, Lin CY. Oestrogen receptors in breast cancer: basic mechanisms and clinical implications. Ecancermedicalscience 2013; 7:370. [PMID: 24222786 PMCID: PMC3816846 DOI: 10.3332/ecancer.2013.370] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Indexed: 12/31/2022] Open
Abstract
Since the discovery of the connection between ovarian hormones and breast cancer, endocrine therapy has been an integral adjuvant treatment for patients with hormone-dependent breast cancers. Oestrogen receptor (ER) plays a central role in mediating the effects of endogenous hormones and therapeutic agents. ER serves as a prognostic marker for responsiveness to endocrine therapy and is targeted either directly by selective oestrogen receptor modulators (SERMs) and pure antagonists or indirectly by aromatase inhibitors (AIs) that block oestrogen production. A significant number of ER-positive patients, however, fail to respond to therapy or develop resistance over time. This review focuses on the current understanding of ER functions and recent advances in genomic technologies and research that have provided a global perspective on hormone and ER activity and led to a number of significant discoveries, including the roles of co-regulatory factors and non-coding RNAs. Mechanistic insights into normal ER functions and therapeutic actions of SERMs and AIs will enable the development of better predictive markers and more effective target mechanisms and ultimately facilitate improvements in disease outcomes and patient survival.
Collapse
Affiliation(s)
- Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204, USA
| | | |
Collapse
|
46
|
Yakimchuk K, Jondal M, Okret S. Estrogen receptor α and β in the normal immune system and in lymphoid malignancies. Mol Cell Endocrinol 2013; 375:121-9. [PMID: 23707618 DOI: 10.1016/j.mce.2013.05.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 05/14/2013] [Accepted: 05/18/2013] [Indexed: 02/07/2023]
Abstract
Estrogens regulate various normal and pathophysiological processes including cancers. Cellular signaling by estrogens is mediated by estrogen receptor α (ERα) and β (ERβ), respectively. Binding of agonists to the ERs affects gene transcription. The main endogenous estrogen, 17β-estradiol (E2), binds to both ERα and ERβ with similar affinity. However, the ligand-binding pocket of ERα and ERβ are slightly different which has allowed the development of selective ER ligands. Importantly, while estrogens via ERα stimulate proliferation, signaling via ERβ inhibits proliferation and promotes apoptosis. In both normal and cancer cells the ERs are co-expressed with ER splice variants which may modify the transcriptional activity of the wild-type receptors. Estrogens have prominent effects on immune functions and both ERα and ERβ are expressed in immune cells and lymphoid malignancies. With regard to lymphoid malignancies, most show estrogen influence as several epidemiological studies of lymphoid cancers demonstrate gender differences in incidence and prognosis with males being more affected. In line with these findings, recent results generated by us have shown that ERβ selective agonists inhibit growth and induce apoptosis in human and murine lymphomas in vivo in xenograft experiments. This suggests that ERβ selective agonists in the future may be useful in the treatment of lymphomas.
Collapse
Affiliation(s)
- Konstantin Yakimchuk
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, SE-141 83 Huddinge, Sweden
| | | | | |
Collapse
|
47
|
Lee MT, Leung YK, Chung I, Tarapore P, Ho SM. Estrogen receptor β (ERβ1) transactivation is differentially modulated by the transcriptional coregulator Tip60 in a cis-acting element-dependent manner. J Biol Chem 2013; 288:25038-25052. [PMID: 23857583 DOI: 10.1074/jbc.m113.476952] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor (ER) β1 and ERα have overlapping and distinct functions despite their common use of estradiol as the physiological ligand. These attributes are explained in part by their differential utilization of coregulators and ligands. Although Tip60 has been shown to interact with both receptors, its regulatory role in ERβ1 transactivation has not been defined. In this study, we found that Tip60 enhances transactivation of ERβ1 at the AP-1 site but suppresses its transcriptional activity at the estrogen-response element (ERE) site in an estradiol-independent manner. However, different estrogenic compounds can modify the Tip60 action. The corepressor activity of Tip60 at the ERE site is abolished by diarylpropionitrile, genistein, equol, and bisphenol A, whereas its coactivation at the AP-1 site is augmented by fulvestrant (ICI 182,780). GRIP1 is an important tethering mediator for ERs at the AP-1 site. We found that coexpression of GRIP1 synergizes the action of Tip60. Although Tip60 is a known acetyltransferase, it is unable to acetylate ERβ1, and its coregulatory functions are independent of its acetylation activity. In addition, we showed the co-occupancy of ERβ1 and Tip60 at ERE and AP-1 sites of ERβ1 target genes. Tip60 differentially regulates the endogenous expression of the target genes by modulating the binding of ERβ1 to the cis-regulatory regions. Thus, we have identified Tip60 as the first dual-function coregulator of ERβ1.
Collapse
Affiliation(s)
- Ming-Tsung Lee
- From the Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health
| | - Yuet-Kin Leung
- From the Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health,; Center for Environmental Genetics, and; Cancer Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267 and
| | - Irving Chung
- From the Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health
| | - Pheruza Tarapore
- From the Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health,; Center for Environmental Genetics, and; Cancer Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267 and
| | - Shuk-Mei Ho
- From the Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health,; Center for Environmental Genetics, and; Cancer Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267 and; the Cincinnati Veteran Affairs Medical Center, Cincinnati, Ohio 45220.
| |
Collapse
|
48
|
Abstract
Despite over 15 years of research, the exact role, if any, played by estrogen receptor β (ERβ) in human breast cancer remains elusive. A large body of data both in vitro and in vivo supports its role as an antiproliferative, pro-apoptotic factor especially when co-expressed with ERα. However, there is a smaller body of data associating ERβ with growth and survival in breast cancer. In clinical studies and most often in cell culture studies, the pro-growth and pro-survival activity of ERβ occurs in ERα-negative breast cancer tissue and cells. This bi-faceted role of ERβ is discussed in this review.
Collapse
Affiliation(s)
- Etienne Leygue
- Department of Biochemistry and Medical Genetics, Manitoba Institute of Cell Biology, University of Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9.
| | | |
Collapse
|
49
|
Estrogen receptor β agonist diarylpropionitrile inhibits lipopolysaccharide-induced regulated on activation normal T cell expressed and secreted (RANTES) production in macrophages by repressing nuclear factor κB activation. Fertil Steril 2013; 100:234-40. [PMID: 23557759 DOI: 10.1016/j.fertnstert.2013.02.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 02/28/2013] [Accepted: 02/28/2013] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To investigate the effect of the estrogen receptor-β (ERβ) agonist diarylpropionitrile (DPN) on lipopolysaccharide (LPS)-induced regulated on activation normal T cell expressed and secreted (RANTES) production in macrophages and the possible mechanisms. DESIGN Cellular and molecular biology experimental study. SETTING University-based research laboratory. PATIENT(S) None. INTERVENTION(S) ERβ mRNA and protein expression determined in murine macrophage cell line RAW264.7 cells using reverse-transcription polymerase chain reaction and Western blot analysis; RANTES production detected by ELISA in LPS-stimulated RAW264.7 cells and ERβ knockdown RAW264.7 cells after the addition of DPN, phosphorylation of p65 and IκB degradation detected by Western blot analysis; and nuclear accumulation of p65 visualized using immunofluorescence. MAIN OUTCOME MEASURE(S) LPS-induced RANTES production and phosphorylation of p65 and IκB. RESULT(S) ERβ was expressed in RAW264.7 cells, and DPN statistically significantly decreased LPS-induced RANTES production in RAW264.7 cells. Small interfering RNA targeting the ERβ gene inhibited the effect of DPN on RANTES production. In addition, DPN inhibited nuclear translocation and phosphorylation of p65 by inhibiting IκB degradation and thus prohibited the activation of nuclear factor κB (NF-κB). CONCLUSION(S) Diarylpropionitrile down-regulates LPS-induced RANTES production via ERβ. This effect of DPN is likely due to repression of nuclear factor κB activation.
Collapse
|
50
|
Estrogen receptor β sustains epithelial differentiation by regulating prolyl hydroxylase 2 transcription. Proc Natl Acad Sci U S A 2013; 110:4708-13. [PMID: 23487784 DOI: 10.1073/pnas.1221654110] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Estrogen receptor β (ERβ) promotes the degradation of hypoxia inducible factor 1α (HIF-1α), which contributes to the ability of this hormone receptor to sustain the differentiation of epithelial and carcinoma cells. Although the loss of ERβ and consequent HIF-1 activation occur in prostate cancer with profound consequences, the mechanism by which ERβ promotes the degradation of HIF-1α is unknown. We report that ERβ regulates the ligand (3β-adiol)-dependent transcription of prolyl hydroxylase 2 (PHD2) also known as Egl nine homolog 1 (EGLN1), a 2-oxoglutarate-dependent dioxygenase that hydroxylates HIF-1α and targets it for recognition by the von Hippel-Lindau tumor suppressor and consequent degradation. ERβ promotes PHD2 transcription by interacting with a unique estrogen response element in the 5' UTR of the PHD2 gene that functions as an enhancer. PHD2 itself is critical for maintaining epithelial differentiation. Loss of PHD2 expression or inhibition of its function results in dedifferentiation with characteristics of an epithelial-mesenchymal transition, and exogenous PHD2 expression in dedifferentiated cells can restore an epithelial phenotype. Moreover, expression of HIF-1α in cells that express PHD2 does not induce dedifferentiation but expression of HIF-1α containing mutations in the proline residues that are hydroxylated by PHD2 induces dedifferentiation. These data describe a unique mechanism for the regulation of HIF-1α stability that involves ERβ-mediated transcriptional regulation of PHD2 and they highlight an unexpected role for PHD2 in maintaining epithelial differentiation.
Collapse
|