1
|
Bu F, Chen L, Sun Y, Zhao B, Wang R. Insight into the Binding Interaction between PEDCs and hERRγ Utilizing Molecular Docking and Molecular Dynamics Simulations. Molecules 2024; 29:3256. [PMID: 39064835 PMCID: PMC11278984 DOI: 10.3390/molecules29143256] [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: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Phenolic environmental endocrine-disrupting chemicals (PEDCs) are persistent EDCs that are widely found in food packaging materials and environmental media and seriously threaten human health and ecological security. Human estrogen-related receptor γ (hERRγ) has been proposed as a mediator for the low-dose effects of many environmental PEDCs; however, the atomic-level descriptions of dynamical structural features and interactions of hERRγ and PEDCs are still unclarified. Herein, how three PEDCs, 4-(1-methylpropyl)phenol (4-sec-butylphenol), 5,6,7,8-tetrahydro-2-naphthol (tetrahydro-2-napthol), and 2,2-bis(4-hydroxy-3,5-dimethoxyphenyl)propane (BP(2,2)(Me)), interact with hERRγ to produce its estrogenic disruption effects was studied. Molecular docking and multiple molecular dynamics (MD) simulations were first conducted to distinguish the detailed interaction pattern of hERRγ with PEDCs. These binding structures revealed that residues around Leu271, Leu309, Leu345, and Phe435 are important when binding with PEDCs. Furthermore, the binding energies of PEDCs with hERRγ were also characterized using the molecular mechanics/Poisson Boltzmann surface area (MM-PBSA) and solvated interaction energy (SIE) methods, and the results showed that the interactions of CH-π, π-π, and hydrogen bonds are the major contributors for hERRγ binding to these three PEDCs. What is striking is that the methoxide groups of BP(2,2)(Me), as hydrophobic groups, can help to reduce the binding energy of PEDCs binding with hERRγ. These results provide important guidance for further understanding the influence of PEDCs on human health problems.
Collapse
Affiliation(s)
- Fanqiang Bu
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161006, China; (F.B.); (Y.S.); (B.Z.)
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lin Chen
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161006, China; (F.B.); (Y.S.); (B.Z.)
| | - Ying Sun
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161006, China; (F.B.); (Y.S.); (B.Z.)
| | - Bing Zhao
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161006, China; (F.B.); (Y.S.); (B.Z.)
- Heilongjiang Provincial Key Laboratory of Surface Active Agent and Auxiliary, Qiqihar University, Qiqihar 161006, China
| | - Ruige Wang
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161006, China; (F.B.); (Y.S.); (B.Z.)
| |
Collapse
|
2
|
Zhang D, Chen H, Wang J, Ji J, Imam M, Zhang Z, Yan S. Current progress and prospects for G protein-coupled estrogen receptor in triple-negative breast cancer. Front Cell Dev Biol 2024; 12:1338448. [PMID: 38476263 PMCID: PMC10928007 DOI: 10.3389/fcell.2024.1338448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a biologically and clinically heterogeneous disease. The G protein-coupled estrogen receptor (GPER) plays a crucial role in mediating the effect of estrogen and estrogen-like compounds in TNBC cells. Compared with other subtypes, GPER has a higher expression in TNBC. The GPER mechanisms have been thoroughly characterized and analyzed in estrogen receptor α (ERα) positive breast cancer, but not in TNBC. Our previous work revealed that a higher expression of GPER mRNA indicates a better prognosis for ERα-positive breast cancer; however, its effects in TNBC differ. Whether GPER could serve as a predictive prognostic marker or therapeutic target for TNBC remains unclear. In this review, we provide a detailed introduction to the subcellular localization of GPER, the different effects of various ligands, and the interactions between GPER and closely associated factors in TNBC. We focused on the internal molecular mechanisms specific to TNBC and thoroughly explored the role of GPER in promoting tumor development. We also discussed the interaction of GPER with specific cytokines and chemokines, and the relationship between GPER and immune evasion. Additionally, we discussed the feasibility of using GPER as a therapeutic target in the context of existing studies. This comprehensive review highlights the effects of GPER on TNBC, providing a framework and directions for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Vanacker JM, Forcet C. ERRα: unraveling its role as a key player in cell migration. Oncogene 2024; 43:379-387. [PMID: 38129506 DOI: 10.1038/s41388-023-02899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023]
Abstract
Cell migration is essential throughout the life of multicellular organisms, and largely depends on the spatial and temporal regulation of cytoskeletal dynamics, cell adhesion and signal transduction. Interestingly, Estrogen-related receptor alpha (ERRα) has been identified as a major regulator of cell migration in both physiological and pathological conditions. ERRα is an orphan member of the nuclear hormone receptor superfamily of transcription factors and displays many biological functions. ERRα is a global regulator of energy metabolism, and it is also highly involved in bone homeostasis, development, differentiation, immunity and cancer progression. Importantly, in some instances, the regulation of these biological processes relies on the ability to orchestrate cell movements. Therefore, this review describes how ERRα-mediated cell migration contributes not only to tissue homeostasis but also to tumorigenesis and metastasis, and highlights the molecular and cellular mechanisms by which ERRα finely controls the cell migratory potential.
Collapse
Affiliation(s)
- Jean-Marc Vanacker
- Centre de Recherche en Cancérologie de Lyon, CNRS UMR5286, Inserm U1052, Université de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, Lyon, France.
| |
Collapse
|
4
|
Chaltel-Lima L, Domínguez F, Domínguez-Ramírez L, Cortes-Hernandez P. The Role of the Estrogen-Related Receptor Alpha (ERRa) in Hypoxia and Its Implications for Cancer Metabolism. Int J Mol Sci 2023; 24:ijms24097983. [PMID: 37175690 PMCID: PMC10178695 DOI: 10.3390/ijms24097983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Under low oxygen conditions (hypoxia), cells activate survival mechanisms including metabolic changes and angiogenesis, which are regulated by HIF-1. The estrogen-related receptor alpha (ERRα) is a transcription factor with important roles in the regulation of cellular metabolism that is overexpressed in hypoxia, suggesting that it plays a role in cell survival in this condition. This review enumerates and analyses the recent evidence that points to the role of ERRα as a regulator of hypoxic genes, both in cooperation with HIF-1 and through HIF-1- independent mechanisms, in invertebrate and vertebrate models and in physiological and pathological scenarios. ERRα's functions during hypoxia include two mechanisms: (1) direct ERRα/HIF-1 interaction, which enhances HIF-1's transcriptional activity; and (2) transcriptional activation by ERRα of genes that are classical HIF-1 targets, such as VEGF or glycolytic enzymes. ERRα is thus gaining recognition for its prominent role in the hypoxia response, both in the presence and absence of HIF-1. In some models, ERRα prepares cells for hypoxia, with important clinical/therapeutic implications.
Collapse
Affiliation(s)
- Leslie Chaltel-Lima
- Segal Cancer Center, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Fabiola Domínguez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco 74360, Mexico
| | - Lenin Domínguez-Ramírez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco 74360, Mexico
| | - Paulina Cortes-Hernandez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco 74360, Mexico
| |
Collapse
|
5
|
Song X, Zhang T, Ding H, Feng Y, Yang W, Yin X, Chen B, Liang Y, Mao Q, Xia W, Yu G, Xu L, Dong G, Jiang F. Non-genetic stratification reveals epigenetic heterogeneity and identifies vulnerabilities of glycolysis addiction in lung adenocarcinoma subtype. Oncogenesis 2022; 11:61. [PMID: 36216804 PMCID: PMC9550819 DOI: 10.1038/s41389-022-00436-0] [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: 04/15/2022] [Revised: 09/12/2022] [Accepted: 09/26/2022] [Indexed: 11/12/2022] Open
Abstract
Lung adenocarcinoma (LUAD) exhibits high heterogeneity and is well known for its high genetic variation. Recently, the understanding of non-genetic variation provides a new perspective to study the heterogeneity of LUAD. Little is known about whether super-enhancers (SEs) may be primarily responsible for the inter-tumor heterogeneity of LUAD. We used super-enhancer RNA (seRNA) levels of a large-scale clinical well-annotated LUAD cohort to stratify patients into three clusters with different prognosis and other malignant characteristics. Mechanistically, estrogen-related receptor alpha (ERRα) in cluster 3-like cell lines acts as a cofactor of BRD4 to assist SE-promoter loops to activate glycolysis-related target gene expression, thereby promoting glycolysis and malignant progression, which confers a therapeutic vulnerability to glycolytic inhibitors. Our study identified three groups of patients according to seRNA levels, among which patients in cluster 3 have the worst prognosis and vulnerability of glycolysis dependency. We also proposed a 3-TF index model to stratify patients with glycolysis-addicted tumors according to tumor SE stratification.
Collapse
Affiliation(s)
- Xuming Song
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Te Zhang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Hanlin Ding
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Yipeng Feng
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Wenmin Yang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Xuewen Yin
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 211198, Nanjing, P. R. China
| | - Bing Chen
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Yingkuan Liang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
| | - Qixing Mao
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
| | - Wenjie Xia
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The affiliated Jiangyin Hospital of Southeast University Medical College, 214400, Jiangyin, P. R. China
| | - Lin Xu
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China. .,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China. .,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China. .,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 211116, Nanjing, P. R. China.
| | - Gaochao Dong
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China. .,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China.
| | - Feng Jiang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 210009, Nanjing, P. R. China. .,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China. .,The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China.
| |
Collapse
|
6
|
Therapeutic Strategies and Potential Actions of Female Sex Steroid Hormones and Their Receptors in Colon Cancer Based on Preclinical Studies. Life (Basel) 2022; 12:life12040605. [PMID: 35455096 PMCID: PMC9032023 DOI: 10.3390/life12040605] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Several epidemiological studies have reported that the use of female sex steroid hormones could reduce the risk of colon cancer (CRC). This review summarizes the available data related to estradiol (E2) and progesterone (P4) single and dual treatments in CRC male and female in vitro and in vivo models, mainly from preclinical studies, alongside their potential molecular mechanisms. Most of the studies showed that E2 exogenous treatment and/or reactivation of its beta receptor (ERβ) significantly inhibited cell proliferation, induced cell cycle arrest, and promoted apoptosis by modulating several molecular pathways. Likewise, the inhibition of ERα receptors produced similar antitumorigenic actions, both in vivo and in vitro, suggesting that E2 could have dual opposing roles in CRC that are dependent on the expression profile of its nuclear receptors. The available studies on P4 are scarce, and the results revealed that in vitro and in vivo treatments with natural and synthetic progesterone were also associated with promising tumoricidal actions. Nevertheless, the combination of E2 with P4 showed enhanced anticancer activities compared with their monotherapy protocols in male–female cell lines and animals. Collectively, the studies suggested that the female sex steroid hormones could provide a novel and effective therapeutic strategy against CRC.
Collapse
|
7
|
Shatnawi A, Ayoub NM, Alkhalifa AE, Ibrahim DR. Estrogen-Related Receptors Gene Expression and Copy Number Alteration Association With the Clinicopathologic Characteristics of Breast Cancer. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234221086713. [PMID: 35359609 PMCID: PMC8961373 DOI: 10.1177/11782234221086713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: It has been suggested that dysregulation of transcription factors expression
or activity plays significant roles in breast cancer (BC) severity and poor
prognosis. Therefore, our study aims to thoroughly evaluate the
estrogen-related receptor isoforms (ESRRs) expression and copy number
alteration (CNA) status and their association with clinicopathologic
characteristics in BC. Methods: A METABRIC dataset consist of 2509 BC patients’ samples was obtained from the
cBioPortal public domain. The gene expression, putative CNA, and relevant
tumor information of ESRRs were retrieved. ESRRs messenger RNA (mRNA)
expression in BC cell lines was obtained from the Cancer Cell Line
Encyclopedia (CCLE). Association and correlation analysis of ESRRs
expression with BC clinicopathologic characteristics and molecular subtype
were performed. Kaplan–Meier survival analysis was conducted to evaluate the
prognostic value of ESRRs expression on patient survival. Results: ESRRα expression correlated negatively with patients’ age and overall
survival, whereas positively correlated with tumor size, the number of
positive lymph nodes, and Nottingham prognostic index (NPI). Conversely,
ESRRγ expression was positively correlated with patients’ age and negatively
correlated with NPI. ESRRα and ESRRγ expression were significantly
associated with tumor grade, expression of hormone receptors, human
epidermal growth factor receptor 2 (HER2), and molecular subtype, whereas
ESRRβ was only associated with tumor stage. A significant and distinct
association of each of ESRRs CNA with various clinicopathologic and
prognostic factors was also observed. Kaplan–Meier survival analysis
demonstrated no significant difference for survival curves among BC patients
with high or low expression of ESRRα, β, or γ. On stratification, high ESRRα
expression significantly reduced survival among premenopausal patients,
patients with grade I/II, and early-stage disease. In BC cell lines, only
ESRRα expression was significantly higher in HER2-positive cells. No
significant association was observed between ESRRβ expression and any of the
clinicopathologic characteristics examined. Conclusions: In this clinical dataset, ESRRα and ESRRγ mRNA expression and CNA show a
significant correlation and association with distinct clinicopathologic and
prognostic parameters known to influence treatment outcomes; however, ESRRβ
failed to show a robust role in BC pathogenesis. ESRRα and ESRRγ can be
employed as therapeutic targets in BC-targeted therapy. However, the role of
ESRRβ in BC pathogenesis remains unclear.
Collapse
Affiliation(s)
- Aymen Shatnawi
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, University of Charleston, Charleston, WV, USA
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Amer E Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Dalia R Ibrahim
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
8
|
Hyldgaard JM, Jensen JB. The Inequality of Females in Bladder Cancer. APMIS 2021; 129:694-699. [PMID: 34582047 DOI: 10.1111/apm.13183] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/10/2021] [Indexed: 11/30/2022]
Abstract
Urinary bladder cancer is worldwide one of the most diagnosed and costly types of cancer. One puzzle in the bladder cancer diagnosis is the disproportional relationship between genders. Males are more likely to be diagnosed with bladder cancer whereas females typically are diagnosed with more adverse disease and worse prognosis, which has led to speculation of the potential role of sex hormones and their receptors in this disease. Estrogen receptors are present in the human bladder, and their role in bladder cancer oncogenesis is increasingly becoming a focus for researchers around the world. This mini-review aims to give a brief overview of the status of female bladder cancer, and to which extend the sex hormones receptors play a role in this. A literature search was performed and included all female original studies on bladder cancer and hormone receptors. Estrogen-receptor alpha seems to be anti-oncogenic whereas estrogen-receptor beta is exhibiting its function pro-oncogenic. The receptor functions may be exercised through mRNA transcriptions and enzymes. Epidemiological studies indicate a potential increase in incidence of bladder cancer for females with earlier age at menopause, and clinical trials are investigating Tamoxifen as a potential treatment in bladder cancer. Increasing evidence supports the theory of bladder cancer development and progression as being partly hormone-dependent. This can lead to a change in conceptual background of bladder cancer etiology and development in the future. Further studies are required to more precise map the use of anti-hormonal drugs in the treatment of this cancer.
Collapse
|
9
|
Tran A, Scholtes C, Songane M, Champagne C, Galarneau L, Levasseur MP, Fodil N, Dufour CR, Giguère V, Saleh M. Estrogen-related receptor alpha (ERRα) is a key regulator of intestinal homeostasis and protects against colitis. Sci Rep 2021; 11:15073. [PMID: 34302001 PMCID: PMC8302669 DOI: 10.1038/s41598-021-94499-5] [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] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
The estrogen-related receptor alpha (ERRα) is a primary regulator of mitochondrial energy metabolism, function and dynamics, and has been implicated in autophagy and immune regulation. ERRα is abundantly expressed in the intestine and in cells of the immune system. However, its role in inflammatory bowel disease (IBD) remains unknown. Here, we report a protective role of ERRα in the intestine. We found that mice deficient in ERRα were susceptible to experimental colitis, exhibiting increased colon inflammation and tissue damage. This phenotype was mediated by impaired compensatory proliferation of intestinal epithelial cells (IEC) following injury, enhanced IEC apoptosis and necrosis and reduced mucus-producing goblet cell counts. Longitudinal analysis of the microbiota demonstrated that loss of ERRα lead to a reduction in microbiome α-diversity and depletion of healthy gut bacterial constituents. Mechanistically, ERRα mediated its protective effects by acting within the radio-resistant compartment of the intestine. It promoted disease tolerance through transcriptional control of key genes involved in intestinal tissue homeostasis and repair. These findings provide new insights on the role of ERRα in the gut and extends our current knowledge of nuclear receptors implicated in IBD.
Collapse
Affiliation(s)
- Allan Tran
- Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Charlotte Scholtes
- Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Mario Songane
- Department of Medicine, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Claudia Champagne
- Department of Medicine, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Luc Galarneau
- Cedars Cancer Centre, Medical Physics, McGill University Health Centre, Montreal, H4A 3J1, Canada
| | - Marie-Pier Levasseur
- Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Biochemistry, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Nassima Fodil
- Department of Biochemistry, McGill University, Montreal, QC, H3A 2B4, Canada
| | | | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Medicine, McGill University, Montreal, QC, H3G 0B1, Canada
- Department of Biochemistry, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Maya Saleh
- Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada.
- Department of Medicine, McGill University, Montreal, QC, H3G 0B1, Canada.
- Department of Life Sciences and Health, CNRS, ImmunoConcEpT, UMR 5164, The University of Bordeaux, 33000, Bordeaux, France.
| |
Collapse
|
10
|
Huang X, Ruan G, Sun P. Estrogen-related receptor alpha copy number variation is associated with ovarian cancer histological grade. J Obstet Gynaecol Res 2021; 47:1878-1883. [PMID: 33751740 DOI: 10.1111/jog.14741] [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: 09/12/2020] [Revised: 01/27/2021] [Accepted: 02/20/2021] [Indexed: 11/27/2022]
Abstract
AIM Copy number variations (CNVs) are related to the genetic and phenotypic diversity of cancers and identifying genetic alterations could improve treatment strategies. Here, we used The Cancer Genome Atlas (TCGA) to explore associations between estrogen-related receptor alpha (ESRRA) CNVs and histological grade in patients with ovarian cancer (OC). METHODS Gene expression data and clinical information of 620 OC patients were obtained from The Cancer Genome Atlas)TCGA and associations between ESRRA CNVs and clinical characteristics were evaluated. Multivariate logistic regression analyses to obtain odds ratios (ORs) using a 95% confidence interval (CI) were performed, adjusting for race, age, histological grade, and tumor size. RESULTS ESRRA CNVs were associated with histological grade (OR 0.6235 [95% CI, 0.3593-0.8877]; p < 0.05) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) CNVs (OR -0.6298 [95% CI, -0.9011 to -0.3585]; p < 0.05). In multivariate analyses, ESRRA CNVs remained significantly associated with histological grade (OR 0.6492 [95% CI, 0.3549-0.9435]; p < 0.05) and PPARGC1A CNVs (OR -0.6236 [95% CI, -0.9269 to 0.3203]; p < 0.05). CONCLUSION There was a significant association between ESRRA CNVs in patients with OC and histological grade of the cancer.
Collapse
Affiliation(s)
- Xiqi Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guanyu Ruan
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Ye S, Xu Y, Wang L, Zhou K, He J, Lu J, Huang Q, Sun P, Wang T. Estrogen-Related Receptor α (ERRα) and G Protein-Coupled Estrogen Receptor (GPER) Synergistically Indicate Poor Prognosis in Patients with Triple-Negative Breast Cancer. Onco Targets Ther 2020; 13:8887-8899. [PMID: 33061416 PMCID: PMC7520096 DOI: 10.2147/ott.s265372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/24/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose The present study aims to demonstrate the correlation between estrogen-related receptor α (ERRα) and G protein-coupled estrogen receptor (GPER) expression and its predictive role in the prognosis of patients with triple-negative breast cancer (TNBC). Methods A retrospective review of 199 cases of TNBC was conducted to assess the GPER and ERRα expression, and its clinicopathologic and prognostic implications. Subsequently, the effects of ERRα and GPER on cell viability, migration, and invasion induced by estrogen were also investigated in vitro. Results Compared to TNBCs with ERRα low expression, ERRα-high patients exhibited higher nuclear grade, more frequent lymph nodal metastasis, a higher rate of local recurrence, and distant metastasis. Survival analyses revealed that ERRα-high patients had decreased overall survival (OS), local recurrence-free survival (LRFS), and distant disease-free survival (DDFS) than ERRα-low patients. The GPER expression level positively correlated with ERRα (R=0.167, P=0.18), and TNBCs with ERRα-low/GPER-low demonstrated the best survival outcomes among groups. In vitro, E2 significantly enhanced cell viability, migration, and invasion in BT-549 and MDA-MB-231 cell lines, which was associated with the increased expression of ERRα. Moreover, the overexpression of ERRα induced by estrogen and G1 (GPER agonist) was reversed by knocking down of GPER and blocking the MAPK signaling with PD98059 in both cell lines. Conclusion Our findings suggest that ERRα and GPER synergistically predict unfavorable prognosis in TNBCs. Mechanically, GPER mediates the upregulation expression of ERRα induced by estrogen and promotes cell viability, migration, and invasion.
Collapse
Affiliation(s)
- Shuang Ye
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, People's Republic of China
| | - Yuanyuan Xu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, People's Republic of China
| | - Ling Wang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, People's Republic of China
| | - Kewen Zhou
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, People's Republic of China
| | - Jiehua He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jiabin Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qitao Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Peng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Tinghuai Wang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, People's Republic of China
| |
Collapse
|
12
|
Zhou Y, Jia Q, Meng X, Chen D, Zhu B. ERRα Regulates OTUB1 Expression to Promote Colorectal Cancer Cell Migration. J Cancer 2019; 10:5812-5819. [PMID: 31737118 PMCID: PMC6843886 DOI: 10.7150/jca.30720] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
Ovarian tumor domain-containing ubiquitin aldehyde binding protein 1 (OTUB1) is overexpressed in many cancers and plays an important role in tumor progression and metastasis. However, the molecular mechanisms underlying OTUB1 overexpression are not clear. In this study, we found that estrogen-related receptor alpha (ERRα, also called NR3B1) binds to OTUB1 promoter and regulates its expression in colorectal cancer. Furthermore, ERRα promoted the migration of CRC cells by inducing vimentin expression via OTUB1. Our data show that OTUB1 is a novel target of ERRα and indicate that ERRα-OTUB1 signaling may play a significant role in CRC metastasis.
Collapse
Affiliation(s)
- Yi Zhou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoqing Meng
- Department of Hematology, the Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Diangang Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
13
|
Ye X, Guo J, Zhang H, Meng Q, Ma Y, Lin R, Yi X, Lu H, Bai X, Cheng J. The enhanced expression of estrogen-related receptor α in human bladder cancer tissues and the effects of estrogen-related receptor α knockdown on bladder cancer cells. J Cell Biochem 2019; 120:13841-13852. [PMID: 30977157 DOI: 10.1002/jcb.28657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 01/15/2023]
Abstract
Estrogen-related receptor α (ERRα) belongs to the superfamily of nuclear orphan receptors. However, the role of ERRα in bladder cancer remains unknown. This study examined the expression of ERRα in bladder cancer tissues and explored the molecular mechanisms of ERRα in bladder cancer progression. The expression of ERRα in bladder cancer tissues from 61 patients was determined by immunohistochemistry. We performed quantitative real-time polymerase chain reaction assay to detect the gene expression levels and carried out Western blot assay to measure protein levels. In vitro functional assays, including colony formation, Cell Counting Kit-8, Transwell invasion, and migration assays, were performed to detect bladder cancer cell growth, proliferation, invasion, and migration, respectively. Flow cytometry was used to determine the cell apoptotic rate of bladder cancer cells. Among the 61 detected bladder cancer tissues, 39 bladder cancer tissues showed positive ERRα immunoreactivity. Higher ERRα immunoreactivity score was significantly associated with TNM stage, tumor grade, distant metastasis, and poor survival in patients with bladder cancer. Univariate and multivariate analyses showed that ERRα immunoreactivity was an independent prognostic factor for overall survival in patients with bladder cancer. ERRα was found to be upregulated in bladder cancer cell lines, and knockdown of ERRα suppressed bladder cancer cell growth, proliferation, invasion, and migration; promoted bladder cancer cell apoptosis; and inhibited the epithelial-mesenchymal transition of bladder cancer cells. On the other hand, bladder cancer cell proliferation, invasion, and migration were significantly enhanced after cells were transfected with an ERRα-overexpressing vector. In vivo tumor growth and metastasis assays showed that ERRα knockdown resulted in remarkable inhibition of tumor growth and tumor metastasis in nude mice. Collectively, our results suggest that the enhanced expression of ERRα may play a key role in the development and progression of bladder cancer and ERRα may serve as an important prognostic factor for bladder cancer.
Collapse
Affiliation(s)
- Xinqing Ye
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jinan Guo
- Department of Urology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen Minimally Invasive Engineering Center, Shenzhen, China
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Hongxiang Zhang
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qinggui Meng
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yun Ma
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Rui Lin
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xianlin Yi
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Haoyuan Lu
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xianzhong Bai
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiwen Cheng
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
14
|
Kokabu T, Mori T, Matsushima H, Yoriki K, Kataoka H, Tarumi Y, Kitawaki J. Antitumor effect of XCT790, an ERRα inverse agonist, on ERα-negative endometrial cancer cells. Cell Oncol (Dordr) 2019; 42:223-235. [PMID: 30706380 DOI: 10.1007/s13402-019-00423-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2019] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The estrogen-related receptor (ERR) α is structurally similar to classical estrogen receptors (ERs), but is considered to be an orphan nuclear receptor. We previously found that ERRα regulates uterine endometrial cancer progression. Here, we investigated the efficacy of XCT790, a selective inverse agonist of ERRα, on endometrial cancer cells in vitro and in vivo. METHODS HEC-1A and KLE, ERα-negative endometrial cancer cells exhibiting high ERRα expression levels, and HEC-1A cell-derived xenograft model mice were treated with XCT790. Transcriptional activity and cell proliferation were examined using luciferase, WST-8 and colony formation assays, respectively. Cell cycle progression was evaluated using flow cytometry, immunofluorescence cytochemistry and Western blotting. Apoptosis was evaluated using a caspase-3/7 activity assay. RESULTS We found that XCT790 significantly inhibited ERRα-induced in vitro transcriptional activity, including that of the vascular endothelial growth factor (VEGF) gene, in a concentration-dependent manner (p < 0.05). We also found that XCT790 suppressed colony formation and cell proliferation in a concentration and time-dependent manner (p < 0.01) without cytotoxicity, and induced apoptosis (p < 0.01). XCT790 was found to cause cell cycle arrest at the mitotic phase. Akt and mTOR phosphorylation was found to be inhibited by XCT790, but PI3K levels were not found to be significantly affected. Combination therapy of XCT790 with paclitaxel elicited a synergistic inhibitory effect. Additionally, we found that XCT790 significantly inhibited in vivo tumor growth and angiogenesis, and induced apoptosis without a reduction in body weight, in xenograft models (p < 0.01). CONCLUSIONS From our data we conclude that XCT790 has an anti-tumor effect on endometrial cancer cells in vitro and in vivo. As such, it may serve as a novel therapeutic agent for endometrial cancer.
Collapse
Affiliation(s)
- Tetsuya Kokabu
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Taisuke Mori
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Hiroshi Matsushima
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kaori Yoriki
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hisashi Kataoka
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yosuke Tarumi
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Jo Kitawaki
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
15
|
Liu G, Sun P, Dong B, Sehouli J. Key regulator of cellular metabolism, estrogen-related receptor α, a new therapeutic target in endocrine-related gynecological tumor. Cancer Manag Res 2018; 10:6887-6895. [PMID: 30588094 PMCID: PMC6296681 DOI: 10.2147/cmar.s182466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The estrogen-related receptor α (ERRα), is an orphan transcription factor. Recently, many studies have reported its regulatory mechanisms and transcriptional targets after identification. Therefore, it may be eligible to join the rank of other nuclear receptors that control almost all aspects of cell metabolism. Cellular metabolism reprogramming plays a key role in fueling malignant change. The purpose of this review was to demonstrate that the ERRα plays an important role in the association between gynecological endocrine-related tumors and energy metabolism. Furthermore, regulation of ERRα may represent a promising strategy to induce cellular metabolic vulnerability of cancer from different origins. Thus, a comprehensive understanding of current treatment strategies may be achieved.
Collapse
Affiliation(s)
- GuiFen Liu
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - PengMing Sun
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China, .,Department of Gynaecology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - BinHua Dong
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - Jalid Sehouli
- Department of Gynaecologic Oncology and Gynaecology, Charité/Campus Virchow-Klinikum, European Competence Centre for Ovarian Cancer University of Berlin, Berlin 13353, Germany
| |
Collapse
|
16
|
Zhou S, Xia H, Xu H, Tang Q, Nie Y, Gong QY, Bi F. ERRα suppression enhances the cytotoxicity of the MEK inhibitor trametinib against colon cancer cells. J Exp Clin Cancer Res 2018; 37:218. [PMID: 30185207 PMCID: PMC6125878 DOI: 10.1186/s13046-018-0862-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/01/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND ERRα, a constitutive transcription factor that regulates energy metabolism, plays an important role in the progression of various tumours. However, its role in cell survival and proliferation and its implication in targeted therapy in colon cancer remains elusive. METHODS The expression of ERRα in colon cancer tissues and cell lines was detected by using western blotting and immunohistochemistry. A wound healing assay and a transwell assay were performed to examine the migration and invasion of the colon cancer cells. A cell viability assay, clonogenic assay, western blot assay and the dual-luciferase reporter assay were employed to study the interaction between trametinib (inhibitor of MEK) and EGF treatment. Flow cytometry, western blotting, quantitative reverse-transcription polymerase chain reaction and xenograft studies were used to identify whether the combination of trametinib and simvastatin had a synergistic effect. RESULTS ERRα positively regulated the cell proliferation, migration and invasion of colon cancer cells, and the suppression of ERRα completely reduced the EGF treatment-induced proliferation of colon cancer cells. Further investigation showed that trametinib partially restrained the up-regulation of ERRα induced by the EGF treatment, and ERRα inhibition increased the sensitivity of colon cancer cells to trametinib. At last, we combined trametinib with simvastatin, a common clinically used drug with a new reported function of transcriptional activity inhibition of ERRα, and found that this combination produced a synergistic effect in inhibiting the proliferation and survival of colon cancer cells in vitro as well as in vivo. CONCLUSIONS The present data indicated that ERRα acted as an oncogene in colon cancer cells, and the combined targeting of ERRα and MEK might be a promising therapeutic strategy for colon cancer treatment.
Collapse
Affiliation(s)
- Sheng Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
- Collaborative Innovation Center for Biotherapy, Sichuan Province, Chengdu, China
| | - Hongwei Xia
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
- Collaborative Innovation Center for Biotherapy, Sichuan Province, Chengdu, China
| | - Huanji Xu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
- Collaborative Innovation Center for Biotherapy, Sichuan Province, Chengdu, China
| | - Qiulin Tang
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
- Collaborative Innovation Center for Biotherapy, Sichuan Province, Chengdu, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digest Diseases, Fourth Military Medical University, Xi’an, Shanxi Province China
| | - Qi yong Gong
- Department of Radiology, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
| | - Feng Bi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Sichuan Province, Chengdu, China
- Laboratory of Molecular Targeted Therapy in Oncology/Department of Medical Oncology, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041 China
| |
Collapse
|
17
|
Zhang J, Guan X, Liang N, Li S. Estrogen-related receptor alpha triggers the proliferation and migration of human non-small cell lung cancer via interleukin-6. Cell Biochem Funct 2018; 36:255-262. [PMID: 29862528 DOI: 10.1002/cbf.3337] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/23/2018] [Accepted: 05/11/2018] [Indexed: 12/27/2022]
Abstract
Human non-small cell lung cancer (NSCLC) is one of the leading causes of cancer deaths worldwide. Estrogenic signals have been suggested to be important for the growth and metastasis of NSCLC cells. Our present data showed that estrogen-related receptor alpha (ERRα), while not ERRβ or ERRγ, was significantly elevated in NSCLC cell lines as compared with that in normal bronchial epithelial cell line BEAS-2B. The expression of ERRα in clinical NSCLC tissues was significantly greater than that in their matched normal adjacent tissues. Over expression of ERRα can trigger the proliferation, migration, and invasion of NSCLC cells, while si-ERRα or ERRα inhibitor showed opposite effects. ERRα can increase the mRNA and protein expression of IL-6, while not IL-8, IL-10, IL-22, VEGF, TGF-β, or TNF-α, in NSCLC cells. Silence of IL-6 attenuated ERRα induced proliferation and cell invasion. Furthermore, our data revealed the inhibition of NF-κB, while not ERK1/2 or PI3K/Akt, abolished ERRα induced production of IL-6. This might be due to that overexpression of ERRα can increase the expression and nuclear translocation of p65 in NSCLC cells. Collectively, our data showed that activation of NF-κB/IL-6 is involved in ERRα induced migration and invasion of NSCLC cells. It suggested that ERRα might be a potential target for NSCLC treatment.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Oncology in First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xiangqian Guan
- Laboratory of the First Affiliated Hospital of University of Science and Technology of China, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Chen H, Pan J, Zhang L, Chen L, Qi H, Zhong M, Shi X, Du J, Li Q. Downregulation of estrogen-related receptor alpha inhibits human cutaneous squamous cell carcinoma cell proliferation and migration by regulating EMT via fibronectin and STAT3 signaling pathways. Eur J Pharmacol 2018; 825:133-142. [PMID: 29476880 DOI: 10.1016/j.ejphar.2018.02.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/09/2018] [Accepted: 02/16/2018] [Indexed: 12/15/2022]
Abstract
Estrogen-related receptor alpha (ERRα), one of orphan nuclear receptors, has been recently revealed as an oncogenic regulator in a variety of cancers. However, the linking gain of ERRα expression and cancer progression in cutaneous squamous cell carcinoma (cSCC) is largely unknown. Here, we showed that the mRNA and protein expression levels of ERRα were markedly higher in A431 cells compared with human keratinocyte cell line HaCaT, and targeted inhibition of ERRα by shRNA or its inverse agonist XCT790 significantly suppressed A431 cells proliferation and migration, while overexpression of ERRα promoted cell proliferation and migration. In addition, the data revealed that ERRα downregulation markedly inhibited the epithelial mesenchymal transition (EMT) of A431 cells with increasing the expression of E-cadherin and decreasing fibronectin (FN) and vimentin. Results from further experiments using Western blot suggested that ERRα suppression inhibited signal transducer and activator of transcription (STAT3) protein expression. In contrast, overexpression of ERRα promoted EMT and the activation of STAT3 pathway. Moreover, treatment with specific STAT3 inhibitor reversed EMT markers in ERRα-overexpressing A431 cells. In tumor xenografts of A431 cells, we further showed that ERRα depletion inhibited cSCC tumor growth in vivo. Taken together, these results demonstrate, for the first time, that ERRα may function as an oncoprotein in cSCC to accelerate tumor aggressiveness by promoting EMT via FN and STAT3 pathway, and it could be a novel target for cSCC therapy.
Collapse
Affiliation(s)
- Haifei Chen
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Jiewen Pan
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Liudi Zhang
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Lu Chen
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China; Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaojin Shi
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China; Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Juan Du
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China; Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
19
|
Liang R, Lin Y, Yuan CL, Liu ZH, Li YQ, Luo XL, Ye JZ, Ye HH. High expression of estrogen-related receptor α is significantly associated with poor prognosis in patients with colorectal cancer. Oncol Lett 2018; 15:5933-5939. [PMID: 29552224 DOI: 10.3892/ol.2018.8011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/21/2017] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of malignancy with high morbidity and mortality rates worldwide. This biologically heterogeneous disease results in diverse therapeutic responses, thus, novel prognostic biomarkers are required to improve CRC treatment. Estrogen-related receptor α (ERRα) is a nuclear orphan receptor, which is associated with estrogen receptor α. The present study aimed to investigate the expression of ERRα in patients with CRC, and explore the association between ERRα expression and clinicopathological factors, local recurrence and prognosis. In the present study, ERRα expression was detected in 15 fresh CRC tissues using quantitative real-time polymerase chain reaction (RT-qPCR) and in 128 paraffin-embedded CRC tissues using immunohistochemistry. The associations between ERRα expression and prognosis of CRC patients were evaluated by univariate, and multivariate (Cox proportional hazards model) analysis. RT-qPCR demonstrated that the mRNA expression of ERRα in CRC tissues was significantly higher compared with that in matched normal tissues. Immunohistochemistry revealed that ERRα high expression was detected in the nuclei of cancer cells from 39.1% (50/128) of CRC tissues. ERRα expression based on immunohistochemical staining was significantly associated with tumor differentiation, tumor invasion, lymph node status and Dukes stage (all P<0.05). Furthermore, patients with high ERRα expression were significantly associated with an increased risk of recurrence and poor prognosis, compared with patients with low ERRα expression. ERRα expression was identified as an independent prognostic factor for patients with CRC. In conclusion, ERRα serves important roles in the progression of CRC and is a potential prognostic factor for patients with CRC.
Collapse
Affiliation(s)
- Rong Liang
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yan Lin
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chun-Ling Yuan
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhi-Hui Liu
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yong-Qiang Li
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiao-Ling Luo
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jia-Zhou Ye
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hai-Hong Ye
- Department of Hepatobilliary Surgery, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530001, P.R. China
| |
Collapse
|
20
|
Ding S, Tang Z, Jiang Y, Huang H, Luo P, Qing B, Zhang S, Tang R. IL-8 Is Involved in Estrogen-Related Receptor α-Regulated Proliferation and Migration of Colorectal Cancer Cells. Dig Dis Sci 2017; 62:3438-3446. [PMID: 28993941 DOI: 10.1007/s10620-017-4779-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 09/21/2017] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS Studies revealed that estrogenic signals were involved in the development of colorectal cancer (CRC), while the roles of estrogen related receptor (ERR) on the progression of CRC have not been well illustrated. Its roles on the development of CRC were investigated. METHODS The expression of ERRα/β/γ in CRC cells were measured. The effects of ERRα on cell proliferation, migration and expression of cytokines were investigated accordingly. RESULTS Our data revealed that the expression of ERRα, while not ERRβ or ERRγ, was significantly increased in CRC cells and clinical CRC tissues. Both the inverse agonist of ERRα (XCT-790) and si-ERRα can inhibit the proliferation of CRC cells. XCT-790 treatment can also suppress the wound healing and in vitro migration of CRC cells. Cytokine assays showed that XCT-790 can significantly decrease the expression of interleukin-8 (IL-8), while not IL-4, IL-6, IL-8, IL-9, IL-10, IL-18, IFN-γ, or TGF-β, in CRC cells. Over expression of ERRα increased the expression of IL-8. Luciferase assay showed XCT-790 decreased the promoter activity of IL-8. XCT-790 can increase the decay of IL-8 mRNA in SW480 cells. The recombinant IL-8 (rIL-8) can rescue XCT-790 induced suppression of proliferation and migration of CRC cells. XCT-790 can decrease the phosphorylation of ERK1/2 and STAT3, two downstream signal molecules of IL-8, in CRC cells. While rIL-8 can markedly attenuate XCT-790 induced dephosphorylation of ERK1/2 and STAT3. CONCLUSION Our data showed that ERRα can trigger the proliferation and migration of CRC cells via up regulation of IL-8. Therefor targeted inhibition of ERRα/IL-8 might be a potential approach for CRC treatment and drug development.
Collapse
Affiliation(s)
- Sijuan Ding
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China
| | - Zhaohui Tang
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China.
| | - Yongjun Jiang
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China
| | - Honglin Huang
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China
| | - Pengfei Luo
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China
| | - Bohua Qing
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China
| | - Siyuan Zhang
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China
| | - Ruoting Tang
- Department of Oncology, The Central Hospital of Yongzhou, No. 396, Yiyun Road, Lengshuitan District, Yongzhou City, 425000, China
| |
Collapse
|
21
|
Carnesecchi J, Cerutti C, Vanacker JM, Forcet C. ERRα protein is stabilized by LSD1 in a demethylation-independent manner. PLoS One 2017; 12:e0188871. [PMID: 29190800 PMCID: PMC5708767 DOI: 10.1371/journal.pone.0188871] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022] Open
Abstract
The LSD1 histone demethylase is highly expressed in breast tumors where it constitutes a factor of poor prognosis and promotes traits of cancer aggressiveness such as cell invasiveness. Recent work has shown that the Estrogen-Related Receptor α (ERRα) induces LSD1 to demethylate the Lys 9 of histone H3. This results in the transcriptional activation of a number of common target genes, several of which being involved in cellular invasion. High expression of ERRα protein is also a factor of poor prognosis in breast tumors. Here we show that, independently of its demethylase activities, LSD1 protects ERRα from ubiquitination, resulting in overexpression of the latter protein. Our data also suggests that the elevation of LSD1 mRNA and protein in breast cancer (as compared to normal tissue) may be a key event to increase ERRα protein, independently of its corresponding mRNA.
Collapse
Affiliation(s)
- Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
22
|
Yun SH, Roh MS, Jeong JS, Park JI. Peroxisome proliferator-activated receptor γ coactivator-1α is a predictor of lymph node metastasis and poor prognosis in human colorectal cancer. Ann Diagn Pathol 2017; 33:11-16. [PMID: 29566941 DOI: 10.1016/j.anndiagpath.2017.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 11/28/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) and PPARγ coactivator-1α (PGC-1α) expression levels are correlated with clinical outcome in breast cancer. However, the potential biological and clinical significance of PPARγ and PGC-1α in colorectal cancer remains unknown. Here we investigated PPARγ and PGC-1α expression in colorectal cancer, and the associations of these expression levels with clinicopathological features. We also evaluated the roles of PPARγ and PGC-1α as prognostic factors in colorectal cancer. We performed immunohistochemical analysis to investigate PPARγ and PGC-1α expression in human colorectal cancer tissues and adjacent normal tissues from 108 primary colorectal cancer patients. We then examined how these expression levels correlated with clinicopathological features. Using the Kaplan-Meier method, we evaluated 3-year disease-free survival (DFS) and overall survival (OS) in patients with tumors expressing different levels of PPARγ and PGC-1α. Our results revealed that PPARγ expression was not significantly correlated with age at surgery, gender, differentiation, depth of infiltration, relapse, or TNM stage. Additionally, PGC-1α expression was not significantly correlated with age at surgery, differentiation, depth of infiltration, relapse, or TNM stage. However, PGC-1α expression was significantly correlated with nodal metastasis (p=0.020). Survival analysis demonstrated reduced OS in the PGC-1α-positive group compared to the PGC-1α-negative group (p=0.03). Our present findings suggest that PGC-1α may be useful for predicting nodal metastasis, and may represent a biomarker for poor prognosis in colorectal cancer.
Collapse
Affiliation(s)
- Seong-Hoon Yun
- Department of Biochemistry, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-Gu, Busan 49201, Republic of Korea
| | - Mee-Sook Roh
- Department of Pathology, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-Gu, Busan 49201, Republic of Korea
| | - Jin-Sook Jeong
- Department of Pathology, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-Gu, Busan 49201, Republic of Korea
| | - Joo-In Park
- Department of Biochemistry, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-Gu, Busan 49201, Republic of Korea.
| |
Collapse
|
23
|
Ning Y, Chen H, Du Y, Ling H, Zhang L, Chen L, Qi H, Shi X, Li Q. A novel compound LingH2-10 inhibits the growth of triple negative breast cancer cells in vitro and in vivo as a selective inverse agonist of estrogen-related receptor α. Biomed Pharmacother 2017; 93:913-922. [PMID: 28715872 DOI: 10.1016/j.biopha.2017.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023] Open
Abstract
Unlike other breast cancer subtypes, targeted therapies for triple negative breast cancer (TNBC) have yet to progress past clinical trial stage to approval. Accumulating evidences demonstrated that expression of estrogen-related receptor alpha (ERRα) indicated worse prognosis and correlated with poor outcome in breast cancers including TNBC. Therefore, ERRα modulators/regulators may be potential in the therapeutic treatment of breast cancers. In the current study, we presented a novel compound LingH2-10 that bound to ERRα, as identified using a time-resolved fluorescence resonance energy transfer assay (TR-FRET) with the IC50 value of 0.64±0.12μM. Further, functional activity was determined by transient transfection luciferase reporter assay in order to validate the utility of the binding affinity in a cellular context. LingH2-10 showed selective inhibition on ERRα transcriptional activity with the IC50 value of 0.58±0.09μM in cell-based luciferase reporter assay. Moreover, representative in vitro results showed that LingH2-10 suppressed the proliferation of various human cancer cells, and inhibited the migration of triple negative breast cancer cell MDA-MB-231. In addition, our results demonstrated that well known ERRα target genes such as PDK4, Osteopontin and pS2, were all significantly down modulated by LingH2-10. In vivo experiments showed that LingH2-10 (30mg/kg, every other day) observably suppressed the growth of MDA-MB-231 xenograft tumors by 42.02% compared to untreated xenograft tumors. Taken together, all these data suggested that LingH2-10, as a selective inverse agonist of ERRα, was a lead compound of anti-cancer agents for treating TNBC patients.
Collapse
Affiliation(s)
- Yang Ning
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China
| | - Haifei Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Yongli Du
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China.
| | - Hao Ling
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China
| | - Liudi Zhang
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Lu Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Huijie Qi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Xiaojin Shi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China; Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qunyi Li
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China; Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
24
|
TRAIL, Wnt, Sonic Hedgehog, TGFβ, and miRNA Signalings Are Potential Targets for Oral Cancer Therapy. Int J Mol Sci 2017; 18:ijms18071523. [PMID: 28708091 PMCID: PMC5536013 DOI: 10.3390/ijms18071523] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/09/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
Clinical studies and cancer cell models emphasize the importance of targeting therapies for oral cancer. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is highly expressed in cancer, and is a selective killing ligand for oral cancer. Signaling proteins in the wingless-type mouse mammary tumor virus (MMTV) integration site family (Wnt), Sonic hedgehog (SHH), and transforming growth factor β (TGFβ) pathways may regulate cell proliferation, migration, and apoptosis. Accordingly, the genes encoding these signaling proteins are potential targets for oral cancer therapy. In this review, we focus on recent advances in targeting therapies for oral cancer and discuss the gene targets within TRAIL, Wnt, SHH, and TGFβ signaling for oral cancer therapies. Oncogenic microRNAs (miRNAs) and tumor suppressor miRNAs targeting the genes encoding these signaling proteins are summarized, and the interactions between Wnt, SHH, TGFβ, and miRNAs are interpreted. With suitable combination treatments, synergistic effects are expected to improve targeting therapies for oral cancer.
Collapse
|
25
|
Chen P, Wang H, Yang F, Chen H, He W, Wang J. Curcumin Promotes Osteosarcoma Cell Death by Activating miR-125a/ERRα Signal Pathway. J Cell Biochem 2017; 118:74-81. [PMID: 27231954 DOI: 10.1002/jcb.25612] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/25/2016] [Indexed: 12/16/2023]
Abstract
Curcumin has demonstrated valuable therapeutic potential against a variety of human cancers including osteosarcoma. However, the molecular mechanisms underlying its anti-tumor effect remain to be poorly understood. By RNA sequence profiling, we found that curcumin significantly down-regulates the expression of estrogen-related receptor alpha (ERRα) in osteosarcoma cells. Overexpression of ERRα diminished curcumin-activated apoptotic cell death and scavenged curcumin-induced reactive oxygen species (ROS), while ERRα silencing sensitized osteosarcoma cells to curcumin, resulting in increased inhibition of cell proliferation. In addition, we found that curcumin suppressed the ERRα gene expression through upregulation of miR-125a. Data from this study revealed a novel mechanism for curcumin-mediated apoptotic cell death, which involves tumor cell killing via activating miR-125a/ERRα pathway. Our studies also provide further support for osteosarcoma therapy by targeting ERRα alone or in combination with curcumin. J. Cell. Biochem. 118: 74-81, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peng Chen
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Guangzhou 510405, People's Republic of China
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California at Davis, 4645 2nd Avenue, Sacramento 95817, California
- Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou 510405, People's Republic of China
| | - Haibin Wang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Guangzhou 510405, People's Republic of China
- Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou 510405, People's Republic of China
| | - Fan Yang
- Shanghai University of Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| | - Hongwu Chen
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California at Davis, 4645 2nd Avenue, Sacramento 95817, California
| | - Wei He
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Guangzhou 510405, People's Republic of China
| | - Junjian Wang
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California at Davis, 4645 2nd Avenue, Sacramento 95817, California
| |
Collapse
|
26
|
The role and impact of estrogens and xenoestrogen on the development of cervical cancer. Biomed Pharmacother 2016; 84:1945-1953. [DOI: 10.1016/j.biopha.2016.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 11/20/2022] Open
|
27
|
Matsushima H, Mori T, Ito F, Yamamoto T, Akiyama M, Kokabu T, Yoriki K, Umemura S, Akashi K, Kitawaki J. Anti-tumor effect of estrogen-related receptor alpha knockdown on uterine endometrial cancer. Oncotarget 2016; 7:34131-48. [PMID: 27153547 PMCID: PMC5085142 DOI: 10.18632/oncotarget.9151] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/16/2016] [Indexed: 12/26/2022] Open
Abstract
Estrogen-related receptor (ERR)α presents structural similarities with estrogen receptor (ER)α. However, it is an orphan receptor not binding to naturally occurring estrogens. This study was designed to investigate the role of ERRα in endometrial cancer progression. Immunohistochemistry analysis on 50 specimens from patients with endometrial cancer showed that ERRα was expressed in all examined tissues and the elevated expression levels of ERRα were associated with advanced clinical stages and serous histological type (p < 0.01 for each). ERRα knockdown with siRNA suppressed angiogenesis via VEGF and cell proliferation in vitro (p < 0.01). Cell cycle and apoptosis assays using flow cytometry and western blot revealed that ERRα knockdown induced cell cycle arrest during the mitotic phase followed by apoptosis initiated by caspase-3. Additionally, ERRα knockdown sensitized cells to paclitaxel. A significant reduction of tumor growth and angiogenesis was also observed in ERRα knockdown xenografts (p < 0.01). These findings indicate that ERRα may serve as a novel molecular target for the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Hiroshi Matsushima
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Taisuke Mori
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Fumitake Ito
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Takuro Yamamoto
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Makoto Akiyama
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Tetsuya Kokabu
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Kaori Yoriki
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Shiori Umemura
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Kyoko Akashi
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| | - Jo Kitawaki
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kajii-cho, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
28
|
Tribollet V, Barenton B, Kroiss A, Vincent S, Zhang L, Forcet C, Cerutti C, Périan S, Allioli N, Samarut J, Vanacker JM. miR-135a Inhibits the Invasion of Cancer Cells via Suppression of ERRα. PLoS One 2016; 11:e0156445. [PMID: 27227989 PMCID: PMC4881992 DOI: 10.1371/journal.pone.0156445] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/13/2016] [Indexed: 01/04/2023] Open
Abstract
MicroRNA-135a (miR-135a) down-modulates parameters of cancer progression and its expression is decreased in metastatic breast cancers (as compared to non-metastatic tumors) as well as in prostate tumors relative to normal tissue. These expression and activity patterns are opposite to those of the Estrogen-Related Receptor α (ERRα), an orphan member of the nuclear receptor family. Indeed high expression of ERRα correlates with poor prognosis in breast and prostate cancers, and the receptor promotes various traits of cancer aggressiveness including cell invasion. Here we show that miR-135a down-regulates the expression of ERRα through specific sequences of its 3'UTR. As a consequence miR-135a also reduces the expression of downstream targets of ERRα. miR-135a also decreases cell invasive potential in an ERRα-dependent manner. Our results suggest that the decreased expression of miR-135a in metastatic tumors leads to elevated ERRα expression, resulting in increased cell invasion capacities.
Collapse
Affiliation(s)
- Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Bruno Barenton
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Auriane Kroiss
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Séverine Vincent
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Ling Zhang
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Séverine Périan
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Nathalie Allioli
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
- Institut des Sciences Pharmaceutiques et Biologiques, Faculté de Pharmacie, Université de Lyon, Lyon, France
| | - Jacques Samarut
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Lyon, France
- UMOMT, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
29
|
Wang D, Wang Y, Liu FQ, Yuan ZY, Mu JJ. High Salt Diet Affects Renal Sodium Excretion and ERRα Expression. Int J Mol Sci 2016; 17:480. [PMID: 27043552 PMCID: PMC4848936 DOI: 10.3390/ijms17040480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 11/18/2022] Open
Abstract
Kidneys regulate the balance of water and sodium and therefore are related to blood pressure. It is unclear whether estrogen-related receptor α (ERRα), an orphan nuclear receptor and transcription factor highly expressed in kidneys, affects the reabsorption of water and sodium. The aim of this study was to determine whether changes in the expressions of ERRα, Na⁺/K⁺-ATPase and epithelial sodium channel (ENaC) proteins affected the reabsorption of water and sodium in kidneys of Dahl salt-sensitive (DS) rats. SS.13BN rats, 98% homologous to the DS rats, were used as a normotensive control group. The 24 h urinary sodium excretion of the DS and SS.13BN rats increased after the 6-week high salt diet intervention, while sodium excretion was increased in DS rats with daidzein (agonist of ERRα) treatment. ERRα expression was decreased, while β- and γ-ENaC mRNA expressions were increased upon high sodium diet treatment in the DS rats. In the chromatin immunoprecipitation (CHIP) assay, positive PCR signals were obtained in samples treated with anti-ERRα antibody. The transcriptional activity of ERRα was decreased upon high salt diet intervention. ERRα reduced the expressions of β- and γ-ENaC by binding to the ENaC promoter, thereby increased Na+ reabsorption. Therefore, ERRα might be one of the factors causing salt-sensitive hypertension.
Collapse
Affiliation(s)
- Dan Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, No. 277 Yanta West Road, Xi'an 710061, China.
| | - Yang Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, No. 277 Yanta West Road, Xi'an 710061, China.
| | - Fu-Qiang Liu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, No. 277 Yanta West Road, Xi'an 710061, China.
| | - Zu-Yi Yuan
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, No. 277 Yanta West Road, Xi'an 710061, China.
| | - Jian-Jun Mu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, No. 277 Yanta West Road, Xi'an 710061, China.
| |
Collapse
|
30
|
Xu Z, Liu J, Gu L, Ma X, Huang B, Pan X. Research progress on the reproductive and non-reproductive endocrine tumors by estrogen-related receptors. J Steroid Biochem Mol Biol 2016; 158:22-30. [PMID: 26802897 DOI: 10.1016/j.jsbmb.2016.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/03/2016] [Accepted: 01/18/2016] [Indexed: 12/20/2022]
Abstract
Oncologists have traditionally considered that tumorigenesis are closely related to classical nuclear estrogen receptors (ERs), such as estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ), through the ligands binding and target gene transcription induction. Estrogen-related receptors (ERRs) have similar structures with ERs, which are also gradually thought to be relevant to reproductive endocrine tumor diseases, even non-reproductive endocrine tumors. In this review, different subtypes of ERRs and their structures firstly will be introduced, then the expression patterns in gynecological oncology (i.e., breast cancer, endometrial cancer, and ovarian cancer), male genitourinary system malignancy especially prostatic cancer along with other non-reproductive endocrine tumors (i.e., lung cancer, colorectal cancer, and liver cancer) will be described, and simultaneously the role of tumorigenesis related to ERRs will be discussed. Therefore, the review is benefit to explore the way of tumor prevention and treatment.
Collapse
Affiliation(s)
- Zhixiang Xu
- Faulty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Jun Liu
- Faulty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Lipeng Gu
- Faulty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Xiaodong Ma
- Faulty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Bin Huang
- Faulty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Xuejun Pan
- Faulty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, Yunnan, China.
| |
Collapse
|
31
|
Tam IS, Giguère V. There and back again: The journey of the estrogen-related receptors in the cancer realm. J Steroid Biochem Mol Biol 2016; 157:13-9. [PMID: 26151739 DOI: 10.1016/j.jsbmb.2015.06.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/09/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022]
Abstract
The identification of two genes encoding polypeptides with structural features common with the estrogen receptor more than a quarter century ago, referred to as the estrogen-related receptors (ERRs), subsequently led to the discovery of several previously unrecognized hormone responsive systems through the application of reverse endocrinology. Paradoxically, the natural ligand(s) associated with members of the ERR subfamily remains to be identified. While initial studies on the mode of action and physiological functions of the ERRs focused on interaction with estrogen signalling in breast cancer, subsequent work showed that the ERRs are ubiquitous master regulators of cellular energy metabolism. This review aims to demonstrate that the ERRs occupy a central node at the interface of cancer and metabolism, and that modulation of their activity may represent a worthwhile strategy to induce metabolic vulnerability in tumors of various origins and thus achieve a more comprehensive response to current therapies.
Collapse
Affiliation(s)
- Ingrid S Tam
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montréal, QC H3A 1A3, Canada
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montréal, QC H3A 1A3, Canada; Departments of Biochemistry, Medicine and Oncology, McGill University, Montréal, PQ H3G 1Y6, Canada.
| |
Collapse
|
32
|
Tiwari A, Swamy S, Gopinath KS, Kumar A. Genomic amplification upregulates estrogen-related receptor alpha and its depletion inhibits oral squamous cell carcinoma tumors in vivo. Sci Rep 2015; 5:17621. [PMID: 26639757 PMCID: PMC4671026 DOI: 10.1038/srep17621] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
The ESRRA gene encodes a transcription factor and regulates several genes, such as WNT11 and OPN, involved in tumorigenesis. It is upregulated in several cancers, including OSCC. We have previously shown that the tumor suppressor miR-125a targets ESRRA, and its downregulation causes upregulation of ESRRA in OSCC. Upregulation of ESRRA in the absence of downregulation of miR-125a in a subset of OSCC samples suggests the involvement of an alternative mechanism. Using TaqMan(®) copy number assay, here we report for the first time that the genomic amplification of ESRRA causes its upregulation in a subset of OSCC samples. Ectopic overexpression of ESRRA led to accelerated cell proliferation, anchorage-independent cell growth and invasion, and inhibited apoptosis. Whereas, knockdown of ESRRA expression by siRNA led to reduced cell proliferation, anchorage-independent cell growth and invasion, and accelerated apoptosis. Furthermore, the delivery of a synthetic biostable ESRRA siRNA to OSCC cells resulted in regression of xenografts in nude mice. Thus, the genomic amplification of ESRRA is another novel mechanism for its upregulation in OSCC. Based on our in vitro and in vivo experiments, we suggest that targeting ESRRA by siRNA could be a novel therapeutic strategy for OSCC and other cancers.
Collapse
Affiliation(s)
- Ankana Tiwari
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Shivananda Swamy
- Department of Surgery, Bangalore Institute of Oncology, Bangalore 560027, India
| | | | - Arun Kumar
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
33
|
Lagerstedt M, Huotari-Orava R, Nyberg R, Mäenpää JU, Snellman E, Laasanen SL. Reduction in ERRα is associated with lichen sclerosus and vulvar squamous cell carcinoma. Gynecol Oncol 2015; 139:536-40. [PMID: 26499936 DOI: 10.1016/j.ygyno.2015.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 10/16/2015] [Accepted: 10/18/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE ERRs (estrogen-related receptors) regulate energy metabolism, the cell cycle and inflammatory processes in both normal and cancer cells. Chronic inflammation induced by lichen sclerosus (LS) or human papilloma virus (HPV) precedes vulvar squamous cell carcinoma (vulvar SCC). We investigated the expression of ERRα, ERRβ and ERRγ in normal vulvar skin, LS as well as LS-dependent and LS-independent/HPV-related vulvar SCC. METHODS A total of 203 samples were analyzed for ERRα, ERRβ and ERRγ by using immunohistochemistry. These included 37 normal vulvar skin samples, 110 LS samples, 6 vulvar intraepithelial neoplasia (VIN) samples and 50 vulvar SCC samples. RESULTS A substantial reduction in or disappearance of ERRα was detected in all vulvar SCC samples. A total of 79% of childhood-onset LS and 51% of adulthood-onset LS lesions showed decreases in ERRα staining. A gradual reduction in ERRα cytoplasmic staining was observed from healthy vulvar skin to precursor lesions and further to SCC. Nuclear ERRα staining was observed in 8/33 (24%) LS-dependent and 10/17 (59%) LS-independent SCC samples. CONCLUSIONS ERRα, a key regulator of cell energy metabolism, may play a role in the pathogenesis of both LS and vulvar SCC.
Collapse
Affiliation(s)
- M Lagerstedt
- Department of Dermatology, Tampere University Hospital, Finland; School of Medicine, University of Tampere, Finland.
| | - R Huotari-Orava
- School of Medicine, University of Tampere, Finland; Fimlab Laboratories, Tampere, Finland
| | - R Nyberg
- Department of Obstetrics and Gynecology, Tampere University Hospital, Finland
| | - J U Mäenpää
- School of Medicine, University of Tampere, Finland; Department of Obstetrics and Gynecology, Tampere University Hospital, Finland
| | - E Snellman
- Department of Dermatology, Tampere University Hospital, Finland; School of Medicine, University of Tampere, Finland
| | - S-L Laasanen
- Department of Dermatology, Tampere University Hospital, Finland
| |
Collapse
|
34
|
Abstract
Cancer is perhaps the fastest growing non-communicable disease in the human population worldwide. Although the molecular mechanism of cancer initiation and progression is known to some extent, however, the majority of pathways responsible for its onset, development and progression are largely unknown. Many members of the nuclear receptors (NRs) superfamily of transcriptional factors have key roles in cancer. Estrogen-related receptor alpha (ERRα) is one of the members of the NR superfamily and studies have linked it with a wide variety of cancers. In endocrine-related cancers such as breast cancer, ERRα regulates a number of target genes directing cell proliferation and growth independent of estrogen receptor alpha (ERα). Knockdown of ERRα in a number of cancer tissues and cell lines significantly reduced tumor growth and malignancy indicating dependence on ERRα activity. The pro-angiogenesis factor vascular endothelial growth factor expression has been shown to be regulated by ERRα and has implications in several types of cancer. The effect of ERRα on cancers seems to be multipronged via regulation of cell cycle regulators, osteopontin, hypoxia inducible factor-1 as well as several energy metabolism genes that are part of glycolysis, TCA cycle, lipogenesis, etc., providing a metabolic twist to cancer. In this article, the action of ERRα on various types of cancers including new developments in this field shall be reviewed.
Collapse
Affiliation(s)
- Harmit S Ranhotra
- a Orphan Nuclear Receptors Laboratory, Department of Biochemistry, St. Edmund's College , Shillong , India
| |
Collapse
|
35
|
Song H, Zhang T, Yang P, Li M, Yang Y, Wang Y, Du J, Pan K, Zhang K. Low doses of bisphenol A stimulate the proliferation of breast cancer cells via ERK1/2/ERRγ signals. Toxicol In Vitro 2015; 30:521-8. [PMID: 26363202 DOI: 10.1016/j.tiv.2015.09.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 08/21/2015] [Accepted: 09/07/2015] [Indexed: 12/20/2022]
Abstract
The effects and mechanisms of bisphenol A (BPA) on the development of breast cancer are still not well illustrated. The present study revealed that nanomolar BPA significantly promoted the proliferation of both estrogen receptor (ER) positive (MCF-7) and negative (SkBr3) breast cancer cells, which was confirmed by up regulation of proliferating cell nuclear antigen (PCNA) and Bcl-2. Neither ERα nor G-protein-coupled estrogen receptor (GPER) mediated this effect of BPA because their inhibitors had no effect on the BPA induced cell proliferation. However, silencing of estrogen related receptor gamma (ERRγ) by its specific siRNA significantly abolished BPA induced proliferation of breast cancer cells, while si-ERRα had no similar effect. Moreover, nanomolar BPA up regulated the mRNA and protein levels of ERRγ and triggered its nuclear translocation via a time dependent manner. Further studies revealed that 10(-8)M BPA obviously increased the phosphorylation of ERK1/2, while had no similar effect on the phosphorylation of JNK and p38 MAPK. Further, PD 98059, the inhibitor of ERK1/2, significantly abolished the BPA induced up regulation of ERRγ and proliferation of breast cancer cells. Collectively, our results revealed that nanomolar BPA can trigger the proliferation of breast cancer cells via ERK1/2/ERRγ signals. Given that nanomolar BPA has been widely detected in human tissues, the clinical relevance of BPA and breast cancer progression should be further investigated.
Collapse
Affiliation(s)
- Haixing Song
- School of Biomedicine Sciences, Chengdu Medical College, Chengdu, China
| | - Tao Zhang
- School of Biomedicine Sciences, Chengdu Medical College, Chengdu, China
| | - Ping Yang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Minhui Li
- Center of Science and Research, Chengdu Medical College, Chengdu, China
| | - Yuhan Yang
- School of Biomedicine Sciences, Chengdu Medical College, Chengdu, China
| | - Yuanyuan Wang
- School of Biomedicine Sciences, Chengdu Medical College, Chengdu, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Kejian Pan
- School of Biomedicine Sciences, Chengdu Medical College, Chengdu, China.
| | - Kun Zhang
- School of Biomedicine Sciences, Chengdu Medical College, Chengdu, China.
| |
Collapse
|
36
|
Ning Z, Du X, Zhang J, Yang K, Miao L, Zhu Y, Yuan H, Wang L, Klocker H, Shi J. PGE2 modulates the transcriptional activity of ERRa in prostate stromal cells. Endocrine 2014; 47:901-12. [PMID: 24760659 DOI: 10.1007/s12020-014-0261-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 03/28/2014] [Indexed: 01/03/2023]
Abstract
The regulation of the transcriptional activity of the estrogen receptor-related receptor a (ERRa) has not yet been clearly documented. Aromatase is a direct target gene of ERRa, and we previously reported that prostaglandin E2 (PGE2) increased the expression of ERRa in the prostate stromal cell line WPMY-1, which ultimately promoted estradiol production by enhancing aromatase gene transcription. Here, we show that PGE2 also affects aromatase expression by regulating ERRa transcriptional activity in prostate stromal cells. When the cells were cultured in serum-free medium, the expression of aromatase was not proportional to the ERRa protein level, if no other stimulation occurred, indicating the absence of a factor that activates ERRa. PGE2 could upregulate aromatase and ERRa response element (ERRE)-reporter expression and also enhance ERRa phosphorylation and nuclear localization. PGE2 functions through the PGE2 receptors (EP) 2 and EP4, which couple to adenylate cyclase. The activation of adenylate cyclase with Forskolin mimicked the PGE2-mediated enhancement of extracellular signal-regulated kinase (ERK) phosphorylation and ERRa target gene expression. Experiments using specific signaling pathway inhibitors showed that both phosphatidylinositol 3-kinase (PI3K) and ERK are involved in ERRa activation, and the PI3K inhibitor was shown to abolish ERK activation. Our results suggest that PGE2 is a modulator of ERRa transcriptional activity. Furthermore, PGE2 activates the EP2/EP4-cAMP-PI3K-ERK signaling pathway, which enhanced ERRa transcriptional potentiality by increasing ERRa phosphorylation and nuclear translocation, subsequently promoting the expression of its target genes, such as aromatase.
Collapse
Affiliation(s)
- Zhaochen Ning
- College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huang JW, Guan BZ, Yin LH, Liu FN, Hu B, Zheng QY, Li FL, Zhong YX, Chen Y. Effects of estrogen-related receptor alpha (ERRα) on proliferation and metastasis of human lung cancer A549 cells. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2014; 34:875-881. [PMID: 25480584 DOI: 10.1007/s11596-014-1367-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/16/2014] [Indexed: 10/24/2022]
Abstract
Estrogen-related receptor alpha (ERRα) plays an important role in the development of hormone-dependent cancers, but its roles in lung cancer remain elusive. The present study was aimed to investigate the effects of ERRα on the proliferation and metastasis of lung cancer A549 cells. The mRNA and protein levels of ERRα were detected in lung cancer A549 and MCF-7 cells and bronchial epithelial BEAS-2B cells by qRT-PCR and Western blotting, respectively. ERRα plasmid transfection and XCT-790 (an inverse agonist of ERRα) were used to up-regulate or down-regulate ERRα expression in A549 cells, respectively. The viability of A549 cells was measured by cell counting kit-8 (CCK-8) and the motility of A549 cells by wound healing assay and Transwell migration/invasion assay. The epithelial markers E-cadherin (E-Cad) and zona occludin-1 (ZO-1), the mesenchymal markers fibronectin (FN) and vimentin (Vim) and the transcription factors (Snail, Zeb1 Twist and Slug) were further detected at mRNA and protein levels by qRT-PCR and Western blotting, respectively. The results showed that ERRα promoted the growth of lung cancer A549 cells in vitro. XCT-790 significantly inhibited the migration and invasion of A549 cells. Over-expression of ERRα promoted the epithelial-to-mesenchymal transition (EMT) of A549 cells, down-regulated the epithelial makers E-Cad and ZO-1, and up-regulated the mesenchymal makers FN and Vim. Silencing of Slug, but not other transcription factors, significantly abolished the ERRα-induced EMT of A549 cells. It was suggested that ERRα promoted the migration and invasion of A549 cells by inducing EMT, and Slug was involved in the process. Targeting ERRα might be an efficient approach for lung cancer treatment.
Collapse
Affiliation(s)
- Jian-Wei Huang
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Bao-Zhang Guan
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Liang-Hong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Fan-Na Liu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Bo Hu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Qi-Yi Zheng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Fo-Lan Li
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Ying-Xue Zhong
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yu Chen
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| |
Collapse
|
38
|
Tiwari A, Shivananda S, Gopinath KS, Kumar A. MicroRNA-125a reduces proliferation and invasion of oral squamous cell carcinoma cells by targeting estrogen-related receptor α: implications for cancer therapeutics. J Biol Chem 2014; 289:32276-32290. [PMID: 25266720 DOI: 10.1074/jbc.m114.584136] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Estrogen-related receptor α (ESRRA) functions as a transcription factor and regulates the expression of several genes, such as WNT11 and OPN. Up-regulation of ESRRA has been reported in several cancers. However, the mechanism underlying its up-regulation is unclear. Furthermore, the reports regarding the role and regulation of ESRRA in oral squamous cell carcinoma (OSCC) are completely lacking. Here, we show that tumor suppressor miR-125a directly binds to the 3'UTR of ESRRA and represses its expression. Overexpression of miR-125a in OSCC cells drastically reduced the level of ESRRA, decreased cell proliferation, and increased apoptosis. Conversely, the delivery of an miR-125a inhibitor to these cells drastically increased the level of ESRRA, increased cell proliferation, and decreased apoptosis. miR-125a-mediated down-regulation of ESRRA impaired anchorage-independent colony formation and invasion of OSCC cells. Reduced cell proliferation and increased apoptosis of OSCC cells were dependent on the presence of the 3'UTR in ESRRA. The delivery of an miR-125a mimic to OSCC cells resulted in marked regression of xenografts in nude mice, whereas the delivery of an miR-125a inhibitor to OSCC cells resulted in a significant increase of xenografts and abrogated the tumor suppressor function of miR-125a. We observed an inverse correlation between the expression levels of miR-125a and ESRRA in OSCC samples. In summary, up-regulation of ESRRA due to down-regulation of miR-125a is not only a novel mechanism for its up-regulation in OSCC, but decreasing the level of ESRRA by using a synthetic miR-125a mimic may have an important role in therapeutic intervention of OSCC and other cancers.
Collapse
Affiliation(s)
- Ankana Tiwari
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012 and
| | - Swamy Shivananda
- Department of Surgery, Bangalore Institute of Oncology, Bangalore 560027, India
| | | | - Arun Kumar
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012 and.
| |
Collapse
|
39
|
Chen P, Wang H, Duan Z, Zou JX, Chen H, He W, Wang J. Estrogen-related receptor alpha confers methotrexate resistance via attenuation of reactive oxygen species production and P53 mediated apoptosis in osteosarcoma cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:616025. [PMID: 24967384 PMCID: PMC4055217 DOI: 10.1155/2014/616025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/25/2014] [Indexed: 01/26/2023]
Abstract
Osteosarcoma (OS) is a malignant tumor mainly occurring in children and adolescents. Methotrexate (MTX), a chemotherapy agent, is widely used in treating OS. However, treatment failures are common due to acquired chemoresistance, for which the underlying molecular mechanisms are still unclear. In this study, we report that overexpression of estrogen-related receptor alpha (ERR α ), an orphan nuclear receptor, promoted cell survival and blocked MTX-induced cell death in U2OS cells. We showed that MTX induced ROS production in MTX-sensitive U2OS cells while ERR α effectively blocked the ROS production and ROS associated cell apoptosis. Our further studies demonstrated that ERR α suppressed ROS induction of tumor suppressor P53 and its target genes NOXA and XAF1 which are mediators of P53-dependent apoptosis. In conclusion, this study demonstrated that ERR α plays an important role in the development of MTX resistance through blocking MTX-induced ROS production and attenuating the activation of p53 mediated apoptosis signaling pathway, and points to ERR α as a novel target for improving osteosarcoma therapy.
Collapse
Affiliation(s)
- Peng Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Haibin Wang
- State Key Hip Joints Center, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 12 Jichang Road, Guangzhou, Guangdong 510405, China
| | - Zhijian Duan
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - June X. Zou
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Hongwu Chen
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Wei He
- State Key Hip Joints Center, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 12 Jichang Road, Guangzhou, Guangdong 510405, China
| | - Junjian Wang
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
40
|
Zou C, Yu S, Xu Z, Wu D, Ng CF, Yao X, Yew DT, Vanacker JM, Chan FL. ERRα augments HIF-1 signalling by directly interacting with HIF-1α in normoxic and hypoxic prostate cancer cells. J Pathol 2014; 233:61-73. [PMID: 24425001 DOI: 10.1002/path.4329] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/09/2013] [Accepted: 01/08/2014] [Indexed: 12/12/2022]
Abstract
Adaptation of cancer cells to a hypoxic microenvironment is important for their facilitated malignant growth and advanced development. One major mechanism mediating the hypoxic response involves up-regulation of hypoxia-inducible factor 1 (HIF-1) expression, which controls reprogramming of energy metabolism and angiogenesis. Oestrogen-related receptor-α (ERRα) is a pivotal regulator of cellular energy metabolism and many biosynthetic pathways, and has also been proposed to be an important factor promoting the Warburg effect in advanced cancer. We and others have previously shown that ERRα expression is increased in prostate cancer and is also a prognostic marker. Here we show that ERRα is oncogenic in prostate cancer and also a key hypoxic growth regulator. ERRα-over-expressing prostate cancer cells were more resistant to hypoxia and showed enhanced HIF-1α protein expression and HIF-1 signalling. These effects could also be observed in ERRα-over-expressing cells grown under normoxia, suggesting that ERRα could function to pre-adapt cancer cells to meet hypoxia stress. Immunoprecipitation and FRET assays indicated that ERRα could physically interact with HIF-1α via its AF-2 domain. A ubiquitination assay showed that this ERRα-HIF-1α interaction could inhibit ubiquitination of HIF-1α and thus reduce its degradation. Such ERRα-HIF-1α interaction could be attenuated by XCT790, an ERRα-specific inverse agonist, resulting in reduced HIF-1α levels. In summary, we show that ERRα can promote the hypoxic growth adaptation of prostate cancer cells via a protective interaction with HIF-1α, suggesting ERRα as a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Chang Zou
- School of Biomedical Sciences, Chinese University of Hong Kong, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lam SSN, Mak ASC, Yam JWP, Cheung ANY, Ngan HYS, Wong AST. Targeting estrogen-related receptor alpha inhibits epithelial-to-mesenchymal transition and stem cell properties of ovarian cancer cells. Mol Ther 2014; 22:743-51. [PMID: 24419103 PMCID: PMC3982489 DOI: 10.1038/mt.2014.1] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/28/2013] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition represents a key event in cancer progression and has emerged as a promising anticancer target. Estrogen-related receptor alpha (ERRα) is frequently elevated in advanced-stage ovarian cancer, but its potential role in tumor progression is not known. Here we show that ERRα functions in epithelial-mesenchymal transition and in subsequent stem cell traits responsible for the acquisition of high degree of aggressiveness and potential for metastasis that are characteristic of ovarian cancer. Importantly, targeted inhibition of ERRα also inhibited the expression of Snail, a repressor of E-cadherin and an inducer of epithelial-mesenchymal transition. Interestingly, induction of Snail resulted from not only changes in mRNA transcription rate but also mRNA stability. We thus identified the miR-200 family as a new player in the ERRα-mediated posttranscriptional regulation of Snail, and antagonism of miR-200a/b could revert the decreased expression of Snail and reversal of epithelial-mesenchymal transition and stem cell characteristics due to ERRα depletion. Finally, we showed that RNA interference-mediated inhibition of ERRα significantly reduced tumor burden, ascites formation, and metastatic peritoneal nodules in vivo in an orthotopic model of ovarian cancer. These results suggest ERRα activation as a mechanism of tumor aggressiveness and imply that targeting ERRα may be a promising approach in ovarian cancer treatment.
Collapse
Affiliation(s)
- Sophia SN Lam
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Abby SC Mak
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Judy WP Yam
- Department of Pathology, University of Hong Kong, Sassoon Road, Pokfulam, Hong Kong
| | - Annie NY Cheung
- Department of Pathology, University of Hong Kong, Sassoon Road, Pokfulam, Hong Kong
| | - Hextan YS Ngan
- Department of Obstetrics and Gynecology, University of Hong Kong, Sassoon Road, Pokfulam, Hong Kong
| | - Alice ST Wong
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| |
Collapse
|
42
|
Zhang JW, Wei YH. Anti-cancer effects of Grailsine-Al-glycoside isolated from Rhizoma Sparganii. Altern Ther Health Med 2014; 14:82. [PMID: 24589031 PMCID: PMC3943449 DOI: 10.1186/1472-6882-14-82] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 02/17/2014] [Indexed: 11/17/2022]
Abstract
Background An embryonic toxicity of Rhizoma sparganii was observed in mice. This study was aimed to evaluate the anticancer effects of Grailsine-Al-glycoside, the bioactive component of Rhizoma sparganii, on estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) cancer cell lines. Methods After A549, HeLa, HepG-2 and MCF-7 cells were treated with Grailsine-Al-glycoside, cell proliferation was analyzed by MTT, cell cycle and apoptosis by flow cytometry, and morphology with an immunofluorescence microscope. Results Grailsine-Al-glycoside strongly suppressed cell proliferation in a dose-dependent fashion in A549, MCF-7, HepG2, and HeLa cells, though this growth inhibitory effect on HepG2 cells was not as strong and long lasting. Compared to the control, Grailsine-Al-glycoside caused a significant increase of apoptosis in A549, MCF-7 and Hela cells. A549 and MCF-7 cells were arrested at the G2/S phase whereas HepG2 cells were arrested at the G1 phase by a high concentration of Grailsine-Al-glycoside . Cell shapes were also changed by the presence of Grailsine-Al-glycoside. Conclusions Grailsine-Al-glycoside from Rhizoma sparganii inhibited the proliferation of ER+ and some ER- cancer cells. Grailsine-Al-glycoside may be used as a chemotherapeutic agent against ER+ and ERRα-expressing ER- cancers.
Collapse
|
43
|
Wang H, Gao M, Wang J. Kaempferol inhibits cancer cell growth by antagonizing estrogen-related receptor α and γ activities. Cell Biol Int 2013; 37:1190-6. [PMID: 23852933 DOI: 10.1002/cbin.10152] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/24/2013] [Indexed: 12/21/2022]
Abstract
Kaempferol is a dietary flavonoid that can function as a selective estrogen receptor modulator (SERM). Estrogen-related receptors alpha and gamma (ERRα and ERRγ) are orphan nuclear receptors that play important roles in mitochondrial biogenesis and cancer development. We have shown that kaempferol can functionally antagonize the activities of ERRs based on both response element reporter systems and target gene analysis. Kaempferol modulation of mitochondrial function and suppression cancer cell growth has been confirmed. These findings suggest that kaempferol may exert their anti-cancer activities through antagonizing ERRs activities.
Collapse
Affiliation(s)
- Haibin Wang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | | | | |
Collapse
|
44
|
Wang H, Wang J. Estrogen-related receptor alpha interacts cooperatively with peroxisome proliferator-activated receptor-gamma coactivator-1alpha to regulate osteocalcin gene expression. Cell Biol Int 2013; 37:1259-65. [PMID: 23788330 DOI: 10.1002/cbin.10148] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/06/2013] [Indexed: 12/24/2022]
Abstract
Osteocalcin is one of the most abundant non-collagenous proteins in bone that is commonly used as a preliminary biomarker in bone formation. Osteocalcin also regulates energy metabolism as a hormone. Estrogen-related receptor alpha (ERRα) is primarily thought to regulate energy homeostasis through interacting with peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α). Intriguingly, ERRα may play a functional role in bone formation. We have found there are 3 ERRα response elements (ERR response element, ERRE) in the osteocalcin promoter, and ERRα interacted cooperatively with PGC-1α could improve the osteocalcin promoter activity, whereas ERRα specific antagonist, XCT-790, inhibits this enhancement. XCT-790 inhibits osteocalcin and ERRα target gene PDKs expression in osteoblast cells. Thus ERRs might have physiological roles in bone formation and energy metabolism through modulating osteocalcin gene expression.
Collapse
Affiliation(s)
- Haibin Wang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | | |
Collapse
|
45
|
Bernatchez G, Giroux V, Lassalle T, Carpentier AC, Rivard N, Carrier JC. ERRα metabolic nuclear receptor controls growth of colon cancer cells. Carcinogenesis 2013; 34:2253-61. [PMID: 23720198 DOI: 10.1093/carcin/bgt180] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The estrogen-related receptor alpha (ERRα) is a nuclear receptor that acts primarily as a regulator of metabolic processes, particularly in tissues subjected to high-energy demand. In addition to its control of energy metabolism and mitochondrial biogenesis, ERRα has recently been associated with cancer progression. Notably, increased expression of ERRα has been shown in several cancerous tissues, including breast, ovary and colon. However, additional studies are required to gain insight into the action of ERRα in cancer biology, particularly in non-endocrine-related cancers. Therefore, using a short hairpin RNA-mediated approach, we investigated whether ERRα is required for the rapid growth of colon cancer cells and to maintain their neoplastic metabolic state. Results show that silencing ERRα significantly impaired colon cancer cell proliferation and colony formation in vitro as well as their in vivo tumorigenic capacity. A pronounced delay in G1-to-S cell cycle phase transition was observed in ERRα-depleted cells in association with reduced cyclin-dependent kinase 2 activity and hyperphosphorylated state of the retinoblastoma protein along with disturbed expression of several cell cycle regulators, including p15 and p27. Interestingly, ERRα-depleted HCT116 cells also displayed significant reduction in expression of a large set of key genes to glycolysis, tricarboxylic acid cycle and lipid synthesis. Furthermore, using (14)C isotope tracer analysis, ERRα depletion in colon cancer cells resulted in reduced glucose incorporation and glucose-mediated lipogenesis in these cells. These findings suggest that ERRα coordinates colon cancer cell proliferation and tumorigenic capacity with energy metabolism. Thus, ERRα could represent a promising therapeutic target in colon cancer.
Collapse
|
46
|
Deblois G, St-Pierre J, Giguère V. The PGC-1/ERR signaling axis in cancer. Oncogene 2013; 32:3483-90. [PMID: 23208510 DOI: 10.1038/onc.2012.529] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/15/2012] [Accepted: 10/15/2012] [Indexed: 12/20/2022]
Abstract
Proliferating cells need to produce a large amount of energy and, at the same time, need to maintain a constant supply of biosynthetic precursors of macromolecules that are used as building blocks for generating new cells. Indeed, many cancer cells undergo a switch from mitochondrial to glycolytic metabolism and display a truncated tricarboxylic acid cycle to match these specific metabolic requirements of proliferation. Understanding the mechanisms by which cancer cells reprogram various metabolic pathways to satisfy their unique bioenergetic requirements has become an active field of research. Concomitantly, it has emerged that members of a family of orphan nuclear receptors known as the estrogen-related receptors (ERRs), working in concert with members of the PPARγ coactivator (PGC)-1 family, act as central transcriptional regulators of metabolic gene networks involved in maintaining energy homeostasis in normal cells. Recent studies have suggested that the PGC-1/ERR transcriptional axis is also important in the metabolic reprogramming of cancer cells. This review focuses on the functional integration of the PGC-1/ERR axis with known oncogenes and the observation that modulation of the activity of this axis can have both pro- and anti-proliferative properties.
Collapse
Affiliation(s)
- G Deblois
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
47
|
Boudjadi S, Bernatchez G, Beaulieu JF, Carrier JC. Control of the human osteopontin promoter by ERRα in colorectal cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:266-76. [PMID: 23680656 DOI: 10.1016/j.ajpath.2013.03.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 02/24/2013] [Accepted: 03/07/2013] [Indexed: 12/19/2022]
Abstract
Colorectal cancer is the second leading cause of death from cancer. Osteopontin (OPN) is a component of tumor extracellular matrix identified as a key marker of cancer progression. The estrogen-related receptor α (ERRα) has been implicated in endocrine-related cancer development and progression, possibly through modulation of cellular energy metabolism. Previous reports that ERRα regulates OPN expression in bone prompted us to investigate whether ERRα controls OPN expression in human colorectal cancer. Using a tissue microarray containing 83 tumor-normal tissue pairs of colorectal cancer samples, we found that tumor epithelial cells displayed higher staining for ERRα than normal mucosa, in correlation with elevated OPN expression. In addition, knocking down endogenous ERRα led to reduced OPN expression in HT29 colon cancer cells. Promoter analysis, inhibition of ERRα activity, and expression and mutation of potential ERRα response elements in the proximal promoter of human OPN showed that ERRα and its obligate co-activator, peroxisome proliferator-activated receptor γ co-activator-1 α, positively control human OPN promoter activity. Furthermore, chromatin immunoprecipitation experiments confirmed in vivo occupancy of the OPN promoter by ERRα in HT29 cells, suggesting that OPN is a direct target of ERRα in colorectal cancer. These findings suggest an additional mechanism by which ERRα participates in the development and progression of colorectal cancer, further supporting the relevance of targeting ERRα with antagonists as anticancer agents.
Collapse
Affiliation(s)
- Salah Boudjadi
- Department of Medicine, Faculty of Medicine and Sciences of Health, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | |
Collapse
|
48
|
Oestrogen receptors are prognostic factors in malignant peritoneal mesothelioma. J Cancer Res Clin Oncol 2013; 139:987-94. [PMID: 23463097 DOI: 10.1007/s00432-013-1408-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/20/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND Malignant peritoneal mesothelioma (MPM) is a rare and fatal cancer. Females are found to survive longer than males after treatment, suggesting a possible involvement of hormonal factors. Estradiol is involved in cellular proliferation of a number of cancers and acts mainly through oestrogen receptors (ERs). Hence, we examined the expression of oestrogen receptors with correlation to prognosis. METHODS Oestrogen receptors expression was examined using immunohistochemistry on 42 paraffin-embedded sections of MPM tumours. Kaplan-Meier survival curves were analysed to determine the significance of ER expression in relation to prognosis. RESULTS ER-β (nuclear) was detected in 33 (79 %) patients. ER-β was also detected in the cellular cytoplasm of 9 (21 %) patients. Presence of ER-β (nuclear) was associated with favourable survival (univariate analysis, P = 0.001), whereas the presence of ER-β (cytoplasm) was associated with a poor survival (P = 0.014). Multivariate Cox regression analysis revealed the absence of ER-β (nuclear) and the presence of ER-β (cytoplasm) to be independent predictive factors for poor disease outcome (hazard ratio 5.4, 95 % confidence interval 1.86-15.75; P = 0.002 and hazard ratio 8.0, 95 % confidence interval 1.8-34; P = 0.005), respectively. ER-α (nuclear) was detected in only 4 (9 %) of patients and not statistically significant (univariate analysis, P = 0.066). CONCLUSION The presence of ER-β (cytoplasm) is associated with poor prognosis. The favourable survival association observed in patients with ER-β (nuclear) raises a question about the molecular mechanisms of the tumorigenic roles of ER-β in each cellular compartment and requires further studies.
Collapse
|
49
|
Felizola SJ, Nakamura Y, Hui XG, Satoh F, Morimoto R, Midorikawa S, Suzuki S, Rainey WE, Sasano H. Estrogen-related receptor α in normal adrenal cortex and adrenocortical tumors: involvement in development and oncogenesis. Mol Cell Endocrinol 2013; 365:207-11. [PMID: 23123734 PMCID: PMC4097865 DOI: 10.1016/j.mce.2012.10.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/16/2012] [Accepted: 10/19/2012] [Indexed: 12/20/2022]
Abstract
AIMS The nuclear hormone receptor estrogen-related receptor α (ERRα) regulates the activation of mitochondrial genes in various human tissues, but its role in the adrenal gland and its disorders has not been defined. Therefore, we examined ERRα expression in both normal adrenal cortex (NAC) and adrenocortical tumor (ACT) in order to study the possible correlation of ERRα with adrenal development and tumor development. METHODS Human adrenal specimens (non-pathological fetal n=7; non-pathological post-birth n=40; aldosterone producing adenoma (APA) n=11; cortisol producing adenoma (CPA) n=11; adrenocortical carcinoma (ACC) n=8) were immunohistochemically examined in this study. NAC (n=13) and ACT (n=28) frozen tissue specimens were also available for studying ERRα mRNA levels. KEY FINDINGS In fetal NAC tissues, ERRα labeling index (LI) in fetal zone (FZ) was significantly higher that that in neocortex (NC), and the differences among age groups for overall mean LI was statistically significant when analyzed according to individual cortical layers. ERRα LI was also significantly higher in ACC than in other types of ACT. ERRα mRNA was detected in NAC and all types of ACT. SIGNIFICANCE Results of our present study suggest a possible role of ERRα in adrenal development and ACC.
Collapse
Affiliation(s)
- Saulo J.A. Felizola
- Tohoku University Graduate School of Medicine, Department of Pathology, Sendai, Japan
| | - Yasuhiro Nakamura
- Tohoku University Graduate School of Medicine, Department of Pathology, Sendai, Japan
| | - Xiao-Gang Hui
- Tohoku University Graduate School of Medicine, Department of Pathology, Sendai, Japan
| | - Fumitoshi Satoh
- Tohoku University Hospital, Division of Nephrology and Hypertension, Sendai, Japan
| | - Ryo Morimoto
- Tohoku University Hospital, Division of Nephrology and Hypertension, Sendai, Japan
| | - Sanae Midorikawa
- Fukushima Medical University, Department of Radiation Health Management, Fukushima, Japan
| | - Shinichi Suzuki
- Fukushima Medical University, Department of Organ Regulatory Surgery, Fukushima, Japan
| | - William E. Rainey
- University of Michigan, Department of Physiology and Medicine, Ann Arbor, Michigan, USA
| | - Hironobu Sasano
- Tohoku University Graduate School of Medicine, Department of Pathology, Sendai, Japan
| |
Collapse
|
50
|
Bianco S, Sailland J, Vanacker JM. ERRs and cancers: effects on metabolism and on proliferation and migration capacities. J Steroid Biochem Mol Biol 2012; 130:180-5. [PMID: 21414406 DOI: 10.1016/j.jsbmb.2011.03.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 01/20/2011] [Accepted: 03/07/2011] [Indexed: 01/06/2023]
Abstract
ERRs are orphan members of the nuclear receptor superfamily which, at least for ERRα and ERRγ display important roles in the control of various metabolic processes. On other hand, correlations have been found between the expression of ERRα and γ and diverse parameters of tumor progression in human cancers. Whereas it is tempting to speculate that ERR receptors act in tumors through the regulation of metabolism, recent data have suggested that they also may directly regulate tumor proliferation and progression independently of their effects on metabolism. The two aspects of tumoral functions of ERR receptors are the purpose of the present review.
Collapse
Affiliation(s)
- Stéphanie Bianco
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364 Lyon cedex 07, France
| | | | | |
Collapse
|