1
|
Tecalco-Cruz AC, López-Canovas L, Azuara-Liceaga E. Estrogen signaling via estrogen receptor alpha and its implications for neurodegeneration associated with Alzheimer's disease in aging women. Metab Brain Dis 2023; 38:783-793. [PMID: 36640216 DOI: 10.1007/s11011-023-01161-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Estrogen receptor alpha (ERα) is a transcription factor activated by estrogenic hormones to regulate gene expression in certain organs, including the brain. In the brain, estrogen signaling pathways are central for maintaining cognitive functions. Herein, we review the neuroprotective effects of estrogens mediated by ERα. The estrogen/ERα pathways are affected by the reduction of estrogens in menopause, and this event may be a risk factor for neurodegeneration associated with Alzheimer's disease in women. Thus, developing a better understanding of estrogen/ERα signaling may be critical for defining new biomarkers and potential therapeutic targets for Alzheimer's disease in women.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Apdo, Postal 03100, Ciudad de México, Mexico.
| | - Lilia López-Canovas
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Apdo, Postal 03100, Ciudad de México, Mexico
| | - Elisa Azuara-Liceaga
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Apdo, Postal 03100, Ciudad de México, Mexico
| |
Collapse
|
2
|
Stossi F, Singh PK, Mistry RM, Johnson HL, Dandekar RD, Mancini MG, Szafran AT, Rao AU, Mancini MA. Quality Control for Single Cell Imaging Analytics Using Endocrine Disruptor-Induced Changes in Estrogen Receptor Expression. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:27008. [PMID: 35167326 PMCID: PMC8846386 DOI: 10.1289/ehp9297] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Diverse toxicants and mixtures that affect hormone responsive cells [endocrine disrupting chemicals (EDCs)] are highly pervasive in the environment and are directly linked to human disease. They often target the nuclear receptor family of transcription factors modulating their levels and activity. Many high-throughput assays have been developed to query such toxicants; however, single-cell analysis of EDC effects on endogenous receptors has been missing, in part due to the lack of quality control metrics to reproducibly measure cell-to-cell variability in responses. OBJECTIVE We began by developing single-cell imaging and informatic workflows to query whether the single cell distribution of the estrogen receptor-α (ER), used as a model system, can be used to measure effects of EDCs in a sensitive and reproducible manner. METHODS We used high-throughput microscopy, coupled with image analytics to measure changes in single cell ER nuclear levels on treatment with ∼100 toxicants, over a large number of biological and technical replicates. RESULTS We developed a two-tiered quality control pipeline for single cell analysis and tested it against a large set of biological replicates, and toxicants from the EPA and Agency for Toxic Substances and Disease Registry lists. We also identified a subset of potentially novel EDCs that were active only on the endogenous ER level and activity as measured by single molecule RNA fluorescence in situ hybridization (RNA FISH). DISCUSSION We demonstrated that the distribution of ER levels per cell, and the changes upon chemical challenges were remarkably stable features; and importantly, these features could be used for quality control and identification of endocrine disruptor toxicants with high sensitivity. When coupled with orthogonal assays, ER single cell distribution is a valuable resource for high-throughput screening of environmental toxicants. https://doi.org/10.1289/EHP9297.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Integrated Microscopy Core, Baylor College of Medicine, Houston, Texas, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
| | - Pankaj K. Singh
- GCC Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
| | - Ragini M. Mistry
- GCC Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
| | - Hannah L. Johnson
- Integrated Microscopy Core, Baylor College of Medicine, Houston, Texas, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
| | | | - Maureen G. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Adam T. Szafran
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Arvind U. Rao
- GCC Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
- Department of Computational Medicine and Bioinformatics, Biostatistics, Biomedical Engineering & Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael A. Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas, USA
- Integrated Microscopy Core, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- GCC Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
| |
Collapse
|
3
|
Calcineurin regulates the stability and activity of estrogen receptor α. Proc Natl Acad Sci U S A 2021; 118:2114258118. [PMID: 34711683 DOI: 10.1073/pnas.2114258118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
Estrogen receptor α (ER-α) mediates estrogen-dependent cancer progression and is expressed in most breast cancer cells. However, the molecular mechanisms underlying the regulation of the cellular abundance and activity of ER-α remain unclear. We here show that the protein phosphatase calcineurin regulates both ER-α stability and activity in human breast cancer cells. Calcineurin depletion or inhibition down-regulated the abundance of ER-α by promoting its polyubiquitination and degradation. Calcineurin inhibition also promoted the binding of ER-α to the E3 ubiquitin ligase E6AP, and calcineurin mediated the dephosphorylation of ER-α at Ser294 in vitro. Moreover, the ER-α (S294A) mutant was more stable and activated the expression of ER-α target genes to a greater extent compared with the wild-type protein, whereas the extents of its interaction with E6AP and polyubiquitination were attenuated. These results suggest that the phosphorylation of ER-α at Ser294 promotes its binding to E6AP and consequent degradation. Calcineurin was also found to be required for the phosphorylation of ER-α at Ser118 by mechanistic target of rapamycin complex 1 and the consequent activation of ER-α in response to β-estradiol treatment. Our study thus indicates that calcineurin controls both the stability and activity of ER-α by regulating its phosphorylation at Ser294 and Ser118 Finally, the expression of the calcineurin A-α gene (PPP3CA) was associated with poor prognosis in ER-α-positive breast cancer patients treated with tamoxifen or other endocrine therapeutic agents. Calcineurin is thus a promising target for the development of therapies for ER-α-positive breast cancer.
Collapse
|
4
|
Rasha F, Sharma M, Pruitt K. Mechanisms of endocrine therapy resistance in breast cancer. Mol Cell Endocrinol 2021; 532:111322. [PMID: 34000350 DOI: 10.1016/j.mce.2021.111322] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/29/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
The most commonly diagnosed breast cancer (BC) subtype is characterized by estrogen receptor (ER) expression. Treatment of this BC subtype typically involves modalities that either suppress the production of estrogen or impede the binding of estrgen to its receptors, constituting the basis for endocrine therapy. While many patients have benefitted from endocrine therapy with clear reduction in mortality and cancer recurrence, one of the clinical hurdles that remain involves overcoming intrinsic (de novo) or acquired resistance to endocrine therapy driven by diverse and complex changes occurring in the tumor microenvironment. Moreover, such resistance may persist even after progression through additional antiestrogen therapies thus demonstrating the importance of further investigation of mechanisms of ER modulation. Here, we discuss a number of advances that provide a better understanding of the complex mechanistic basis for resistance to endocrine therapy as well as future therapeutic maneuvers that may break this resistance.
Collapse
Affiliation(s)
- Fahmida Rasha
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA
| | - Monica Sharma
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA
| | - Kevin Pruitt
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA.
| |
Collapse
|
5
|
Jacquemetton J, Kassem L, Poulard C, Dahmani A, De Plater L, Montaudon E, Sourd L, Morisset L, El Botty R, Chateau-Joubert S, Vacher S, Bièche I, Treilleux I, Trédan O, Marangoni E, Le Romancer M. Analysis of genomic and non-genomic signaling of estrogen receptor in PDX models of breast cancer treated with a combination of the PI3K inhibitor alpelisib (BYL719) and fulvestrant. Breast Cancer Res 2021; 23:57. [PMID: 34020697 PMCID: PMC8139055 DOI: 10.1186/s13058-021-01433-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Endocrine therapies targeting estrogen signaling have significantly improved breast cancer (BC) patient survival, although 40% of ERα-positive BCs do not respond to those therapies. Aside from genomic signaling, estrogen triggers non-genomic pathways by forming a complex containing methylERα/Src/PI3K, a hallmark of aggressiveness and resistance to tamoxifen. We aimed to confirm the prognostic value of this complex and investigated whether its targeting could improve tumor response in vivo. METHODS The interaction of ERα/Src and ERα/PI3K was studied by proximity ligation assay (PLA) in a cohort of 440 BC patients. We then treated patient-derived BC xenografts (PDXs) with fulvestrant or the PI3K inhibitor alpelisib (BYL719) alone or in combination. We analyzed their anti-proliferative effects on 6 ERα+ and 3 ERα- PDX models. Genomic and non-genomic estrogen signaling were assessed by measuring ERα/PI3K interaction by PLA and the expression of estrogen target genes by RT-QPCR, respectively. RESULTS We confirmed that ERα/Src and ERα/PI3K interactions were associated with a trend to poorer survival, the latter displaying the most significant effects. In ERα+ tumors, the combination of BYL719 and fulvestrant was more effective than fulvestrant alone in 3 models, irrespective of PI3K, PTEN status, or ERα/PI3K targeting. Remarkably, resistance to fulvestrant was associated with non-genomic ERα signaling, since genomic degradation of ERα was unaltered in these tumors, whereas the treatment did not diminish the level of ERα/PI3K interaction. Interestingly, in 2 ERα- models, fulvestrant alone impacted tumor growth, and this was associated with a decrease in ERα/PI3K interaction. CONCLUSIONS Our results demonstrate that ERα/PI3K may constitute a new prognostic marker, as well as a new target in BC. Indeed, resistance to fulvestrant in ERα+ tumors was associated with a lack of impairment of ERα/PI3K interaction in the cytoplasm. In addition, an efficient targeting of ERα/PI3K in ERα- tumors could constitute a promising therapeutic option.
Collapse
Affiliation(s)
- Julien Jacquemetton
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Loay Kassem
- Clinical Oncology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Coralie Poulard
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France
| | - Ahmed Dahmani
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Ludmilla De Plater
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Elodie Montaudon
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Laura Sourd
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Ludivine Morisset
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Rania El Botty
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Sophie Chateau-Joubert
- École Nationale Vétérinaire d'Alfort, BioPôle Alfort, 94704, Maisons-Alfort Cedex, France
| | | | - Ivan Bièche
- Genetics Department, Institut Curie, Paris, France
| | - Isabelle Treilleux
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,Pathology Department, Centre Léon Bérard, F-69000, Lyon, France
| | - Olivier Trédan
- Université de Lyon, F-69000, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France.,Medical Oncology Department, Centre Léon Bérard, F-69000, Lyon, France
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Muriel Le Romancer
- Université de Lyon, F-69000, Lyon, France. .,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France. .,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000, Lyon, France. .,Centre de Recherche en Cancérologie de Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Bâtiment D, 28 rue Laennec, 69373, Lyon Cedex 08, France.
| |
Collapse
|
6
|
Zhu J, Lv Y, Hao J, Shi T, Wang S, Wang K, Fan X, Guo Y, Zhang J, Li J. N-myc downstream-regulated gene 2 promotes the protein stability of estrogen receptor beta via inhibition of ubiquitin-protein ligase E3A to suppress colorectal cancer. J Gastrointest Oncol 2020; 11:1200-1213. [PMID: 33456993 DOI: 10.21037/jgo-20-557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background N-myc downstream-regulated gene 2 (NDRG2) and estrogen receptor beta (ERβ) both play key roles in cellular differentiation in colorectal cancer (CRC). Previous studies have demonstrated that ERβ co-locates with and directly transactivates NDRG2. However, the effect of NDRG2 on ERβ and its underlying mechanism remain largely unknown. Our aim of the study is to explore the effect of NDRG2 on ERβ and their contributions to progression of CRC. Methods The Cancer Genome Atlas (TCGA) database was first utilized to validate the clinical significance of ERβ and NDRG2 in CRC. MTT and scratch migration assays were carried out to verify the role of ERβ and NDRG2 in CRC cells. Western blotting and polymerase chain reaction were performed to analyze the effect of NDRG2 on ERβ, and an immunoprecipitation assay was conducted to explore the protein-protein interaction. Results ERβ and NDRG2 were both found to be significantly down-regulated in tumor tissues from the TCGA-CRC database. NDRG2 was also observed to enhance the protein stability of ERβ while could not change messenger RNA (mRNA) level of ESR2 (encoding ERβ). A positive relationship was found to exist between the two proteins in CRC cells, with NDRG2 prolonging the half-life of ERβ and improving its nuclear translocation. Through detecting expression of ERβ downstream genes (such as TP53 and JNK) and performing related function experiment, we demonstrated that NDRG2 could promote transcriptional activation of ERβ target genes and enhance the function of tumor suppressors when the ERβ agonist diarylpropionitrile (DPN). The immunoprecipitation assay showed that NDRG2 could affect the complex components of ubiquitin-protein ligase E3A (UBE3A, known as E6AP) and ERβ, reducing the ubiquitin-mediated proteasome degradation of ERβ. Conclusions In the current study, we found that NDRG2 could bind with UBE3A to hinder the binding of UBE3A with ERβ. Moreover, a positive feedback loop was discovered between NDRG2 and ERβ, which provides a novel insight and therapeutic target for CRC.
Collapse
Affiliation(s)
- Jun Zhu
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yongzhi Lv
- The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jun Hao
- Department of Experiment Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Tingyu Shi
- Department of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Shuai Wang
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ke Wang
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaoyan Fan
- Department of Experiment Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuan Guo
- School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Jian Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, China
| | - Jipeng Li
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
7
|
Shagufta, Ahmad I, Mathew S, Rahman S. Recent progress in selective estrogen receptor downregulators (SERDs) for the treatment of breast cancer. RSC Med Chem 2020; 11:438-454. [PMID: 33479648 PMCID: PMC7580774 DOI: 10.1039/c9md00570f] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
Selective estrogen receptor downregulators (SERDs) are a novel class of compounds capable of reducing the ERα protein level and blocking ER activity. Therefore, SERDs are considered as a significant therapeutic approach to treat ER+ breast cancer in both early stage and more advanced drug-resistant cases. After the FDA approval of a steroidal drug, fulvestrant, as a SERD for the treatment of breast cancer in patients who have progressed on antihormonal agents, several molecules with diverse chemical structures have been rapidly developed, studied and evaluated for selective estrogen receptor downregulation activity. Here we compile the promising SERDs reported in recent years and discuss the chemical structure and pharmacological profile of the most potent compound of the considered series. Because of the availability of only a limited number of effective drugs for the treatment of breast cancer, the quest for a potent SERD with respectable activity and bioavailability is still ongoing. The goal of this article is to make available to the reader an overview of the current progress in SERDs and provide clues for the future discovery and development of novel pharmacological potent SERDs for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shagufta
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates . ;
| | - Irshad Ahmad
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates . ;
| | - Shimy Mathew
- Department of Biotechnology , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates
| | - Sofia Rahman
- Department of Biotechnology , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates
| |
Collapse
|
8
|
Ohgane K, Yoshioka H, Hashimoto Y. Multiplexing fluorogenic esterase-based viability assay with luciferase assays. MethodsX 2019; 6:2013-2020. [PMID: 31667098 PMCID: PMC6812399 DOI: 10.1016/j.mex.2019.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/10/2019] [Indexed: 11/24/2022] Open
Abstract
Luciferase-based reporter assays are one of the most common cell-based screening formats for drug discovery, and simultaneous evaluation of the cytotoxic effect of test compounds is of great value in reducing false-positives. Here we share a multiplex assay protocol that allows sequential measurement of cell viability (cell number) and luciferase activity of the same sample in a multi-well-plate format. The viability assay employs a fluorogenic esterase substrate, CytoRed. This protocol allows sequential measurement of endogenous esterase activity (as a surrogate for cell number) and then luciferase activity in a single sample. The protocol eliminates the need for parallel viability assay or protein assay using separate aliquots of the lysate. This protocol is especially useful for assays with cells stably expressing a luciferase construct, for which co-transfection of another reporter gene is not a viable option.
Collapse
Affiliation(s)
- Kenji Ohgane
- Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hiromasa Yoshioka
- Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Yuichi Hashimoto
- Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| |
Collapse
|
9
|
Cairns J, Ingle JN, Kalari KR, Shepherd LE, Kubo M, Goetz MP, Weinshilboum RM, Wang L. The lncRNA MIR2052HG regulates ERα levels and aromatase inhibitor resistance through LMTK3 by recruiting EGR1. Breast Cancer Res 2019; 21:47. [PMID: 30944027 PMCID: PMC6448248 DOI: 10.1186/s13058-019-1130-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/25/2019] [Indexed: 01/10/2023] Open
Abstract
Background Our previous genome-wide association study using the MA.27 aromatase inhibitors adjuvant trial identified SNPs in the long noncoding RNA MIR2052HG associated with breast cancer-free interval. MIR2052HG maintained ERα both by promoting AKT/FOXO3-mediated ESR1 transcription and by limiting ubiquitin-mediated ERα degradation. Our goal was to further elucidate MIR2052HG’s mechanism of action. Methods RNA-binding protein immunoprecipitation assays were performed to demonstrate that the transcription factor, early growth response protein 1 (EGR1), worked together with MIR2052HG to regulate that lemur tyrosine kinase-3 (LMTK3) transcription in MCF7/AC1 and CAMA-1 cells. The location of EGR1 on the LMTK3 gene locus was mapped using chromatin immunoprecipitation assays. The co-localization of MIR2052HG RNA and the LMTK3 gene locus was determined using RNA-DNA dual fluorescent in situ hybridization. Single-nucleotide polymorphisms (SNP) effects were evaluated using a panel of human lymphoblastoid cell lines. Results MIR2052HG depletion in breast cancer cells results in a decrease in LMTK3 expression and cell growth. Mechanistically, MIR2052HG interacts with EGR1 and facilitates its recruitment to the LMTK3 promoter. LMTK3 sustains ERα levels by reducing protein kinase C (PKC) activity, resulting in increased ESR1 transcription mediated through AKT/FOXO3 and reduced ERα degradation mediated by the PKC/MEK/ERK/RSK1 pathway. MIR2052HG regulated LMTK3 in a SNP- and aromatase inhibitor-dependent fashion: the variant SNP increased EGR1 binding to LMTK3 promoter in response to androstenedione, relative to wild-type genotype, a pattern that can be reversed by aromatase inhibitor treatment. Finally, LMTK3 overexpression abolished the effect of MIR2052HG on PKC activity and ERα levels. Conclusions Our findings support a model in which the MIR2052HG regulates LMTK3 via EGR1, and LMTK3 regulates ERα stability via the PKC/MEK/ERK/RSK1 axis. These results reveal a direct role of MIR2052HG in LMTK3 regulation and raise the possibilities of targeting MIR2052HG or LMTK3 in ERα-positive breast cancer. Electronic supplementary material The online version of this article (10.1186/s13058-019-1130-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junmei Cairns
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - James N Ingle
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lois E Shepherd
- NCIC Clinical Trials Group, Kingston, Ontario, K7L 3N6, Canada
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama City, 230-0045, Japan
| | - Matthew P Goetz
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
10
|
Tecalco-Cruz AC, Ramírez-Jarquín JO, Cruz-Ramos E. Estrogen Receptor Alpha and its Ubiquitination in Breast Cancer Cells. Curr Drug Targets 2019; 20:690-704. [DOI: 10.2174/1389450119666181015114041] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 12/23/2022]
Abstract
More than 70% of all breast cancer cases are estrogen receptor alpha-positive (ERα). ERα is a member of the nuclear receptor family, and its activity is implicated in the gene transcription linked to the proliferation of breast cancer cells, as well as in extranuclear signaling pathways related to the development of resistance to endocrine therapy. Protein-protein interactions and posttranslational modifications of ERα underlie critical mechanisms that modulate its activity. In this review, the relationship between ERα and ubiquitin protein (Ub), was investigated in the context of breast cancer cells. Interestingly, Ub can bind covalently or non-covalently to ERα resulting in either a proteolytic or non-proteolytic fate for this receptor. Thereby, Ub-dependent molecular pathways that modulate ERα signaling may play a central role in breast cancer progression, and consequently, present critical targets for treatment of this disease.
Collapse
Affiliation(s)
- Angeles C. Tecalco-Cruz
- Instituto de Investigaciones Biomedicas. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| | - Josué O. Ramírez-Jarquín
- Instituto de Fisiologia Celular. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| | - Eduardo Cruz-Ramos
- Instituto de Investigaciones Biomedicas. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| |
Collapse
|
11
|
Hu J, Hu B, Wang M, Xu F, Miao B, Yang CY, Wang M, Liu Z, Hayes DF, Chinnaswamy K, Delproposto J, Stuckey J, Wang S. Discovery of ERD-308 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Receptor (ER). J Med Chem 2019; 62:1420-1442. [DOI: 10.1021/acs.jmedchem.8b01572] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
12
|
Nagasawa J, Govek S, Kahraman M, Lai A, Bonnefous C, Douglas K, Sensintaffar J, Lu N, Lee K, Aparicio A, Kaufman J, Qian J, Shao G, Prudente R, Joseph JD, Darimont B, Brigham D, Maheu K, Heyman R, Rix PJ, Hager JH, Smith ND. Identification of an Orally Bioavailable Chromene-Based Selective Estrogen Receptor Degrader (SERD) That Demonstrates Robust Activity in a Model of Tamoxifen-Resistant Breast Cancer. J Med Chem 2018; 61:7917-7928. [PMID: 30086626 DOI: 10.1021/acs.jmedchem.8b00921] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
About 75% of breast cancers are estrogen receptor alpha (ER-α) positive, and women typically initially respond well to antihormonal therapies such as tamoxifen and aromatase inhibitors, but resistance often emerges. Fulvestrant is a steroid-based, selective estrogen receptor degrader (SERD) that both antagonizes and degrades ER-α and shows some activity in patients who have progressed on antihormonal agents. However, fulvestrant must be administered by intramuscular injections that limit its efficacy. We describe the optimization of ER-α degradation efficacy of a chromene series of ER modulators resulting in highly potent and efficacious SERDs such as 14n. When examined in a xenograft model of tamoxifen-resistant breast cancer, 14n (ER-α degradation efficacy = 91%) demonstrated robust activity, while, despite superior oral exposure, 15g (ER-α degradation efficacy = 82%) was essentially inactive. This result suggests that optimizing ER-α degradation efficacy in the MCF-7 cell line leads to compounds with robust effects in models of tamoxifen-resistant breast cancer derived from an MCF-7 background.
Collapse
|
13
|
Tecalco-Cruz AC, Ramírez-Jarquín JO. Polyubiquitination inhibition of estrogen receptor alpha and its implications in breast cancer. World J Clin Oncol 2018; 9:60-70. [PMID: 30148069 PMCID: PMC6107474 DOI: 10.5306/wjco.v9.i4.60] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/22/2018] [Accepted: 06/28/2018] [Indexed: 02/06/2023] Open
Abstract
Estrogen receptor alpha (ERα) is detected in more than 70% of the cases of breast cancer. Nuclear activity of ERα, a transcriptional regulator, is linked to the development of mammary tumors, whereas the extranuclear activity of ERα is related to endocrine therapy resistance. ERα polyubiquitination is induced by the estradiol hormone, and also by selective estrogen receptor degraders, resulting in ERα degradation via the ubiquitin proteasome system. Moreover, polyubiquitination is related to the ERα transcription cycle, and some E3-ubiquitin ligases also function as coactivators for ERα. Several studies have demonstrated that ERα polyubiquitination is inhibited by multiple mechanisms that include posttranslational modifications, interactions with coregulators, and formation of specific protein complexes with ERα. These events are responsible for an increase in ERα protein levels and deregulation of its signaling in breast cancers. Thus, ERα polyubiquitination inhibition may be a key factor in the progression of breast cancer and resistance to endocrine therapy.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Programa de Investigación de Cáncer de Mama (PICM), Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México 04510, México
| | - Josué O Ramírez-Jarquín
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México 04510, México
| |
Collapse
|
14
|
Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol Ther 2018; 186:1-24. [DOI: 10.1016/j.pharmthera.2017.12.012] [Citation(s) in RCA: 319] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Nucleo-cytoplasmic transport of estrogen receptor alpha in breast cancer cells. Cell Signal 2017; 34:121-132. [PMID: 28341599 DOI: 10.1016/j.cellsig.2017.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/19/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023]
Abstract
Approximately 70% cases of breast cancers exhibit high expression and activity levels of estrogen receptor alpha (ERα), a transcription regulator that induces the expression of genes associated with cellular proliferation and survival. These nuclear functions of the receptor are associated with the development of breast cancer. However, ERα localization is not static, but rather, dynamic with continuous shuttling between the nucleus and the cytoplasm. Interestingly, both the nuclear import and export of ERα are modulated by several stimuli that include estradiol, antiestrogens, and growth factors. As ERα nuclear accumulation is critical to the regulation of gene expression, nuclear export of this receptor modulates the intensity and duration of its transcriptional activity. Thus, the subcellular spatial distribution of ERα ensures tight modulation of its concentration in cellular compartments, as well as of its nuclear and extranuclear functions. In this review, we will discuss current findings regarding the biological importance of molecular mechanisms of, and proteins responsible for, the nuclear import and export of ERα in breast cancer cells.
Collapse
|
16
|
Traboulsi T, El Ezzy M, Gleason JL, Mader S. Antiestrogens: structure-activity relationships and use in breast cancer treatment. J Mol Endocrinol 2017; 58:R15-R31. [PMID: 27729460 PMCID: PMC5148801 DOI: 10.1530/jme-16-0024] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022]
Abstract
About 70% of breast tumors express estrogen receptor alpha (ERα), which mediates the proliferative effects of estrogens on breast epithelial cells, and are candidates for treatment with antiestrogens, steroidal or non-steroidal molecules designed to compete with estrogens and antagonize ERs. The variable patterns of activity of antiestrogens (AEs) in estrogen target tissues and the lack of systematic cross-resistance between different types of molecules have provided evidence for different mechanisms of action. AEs are typically classified as selective estrogen receptor modulators (SERMs), which display tissue-specific partial agonist activity (e.g. tamoxifen and raloxifene), or as pure AEs (e.g. fulvestrant), which enhance ERα post-translational modification by ubiquitin-like molecules and accelerate its proteasomal degradation. Characterization of second- and third-generation AEs, however, suggests the induction of diverse ERα structural conformations, resulting in variable degrees of receptor downregulation and different patterns of systemic properties in animal models and in the clinic.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/chemistry
- Antineoplastic Agents, Hormonal/pharmacology
- Antineoplastic Agents, Hormonal/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Clinical Trials as Topic
- Drug Evaluation, Preclinical
- Drug Resistance, Neoplasm
- Estrogen Antagonists/chemistry
- Estrogen Antagonists/pharmacology
- Estrogen Antagonists/therapeutic use
- Estrogen Receptor alpha/antagonists & inhibitors
- Estrogen Receptor alpha/chemistry
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Models, Molecular
- Molecular Conformation
- Molecular Structure
- Mutation
- Protein Binding
- Protein Processing, Post-Translational
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/chemistry
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Selective Estrogen Receptor Modulators/chemistry
- Selective Estrogen Receptor Modulators/pharmacology
- Selective Estrogen Receptor Modulators/therapeutic use
- Structure-Activity Relationship
- Treatment Outcome
Collapse
Affiliation(s)
- T Traboulsi
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
- Department of Biochemistry and Molecular MedicineUniversité de Montréal, Montréal, Québec, Canada
| | - M El Ezzy
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
| | - J L Gleason
- Department of ChemistryMcGill University, Montréal, Québec, Canada
| | - S Mader
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
- Department of Biochemistry and Molecular MedicineUniversité de Montréal, Montréal, Québec, Canada
| |
Collapse
|
17
|
Bogliolo S, Cassani C, Dominoni M, Orlandini A, Ferrero S, Iacobone AD, Viazzo F, Venturini PL, Spinillo A, Gardella B. The role of fulvestrant in endometrial cancer. Expert Opin Drug Metab Toxicol 2016; 13:537-544. [PMID: 27696906 DOI: 10.1080/17425255.2016.1244264] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Endometrial cancer is the most common malignancy of the female genital tract in industrialized countries. The traditional treatment of endometrial cancer is based on a surgical approach. In recent years, systemic endocrine therapy has demonstrated good efficacy in recurrent or metastatic setting, delaying progression, ameliorating quality of life and palliating symptoms. Areas covered: Phase I and II studies on selective estrogen receptor down-regulators used for the treatment of endometrial cancer treatment have been reviewed. The pharmacokinetic and pharmacodynamic features of selective receptor down-regulators have been also investigated. Expert opinion: Selective estrogen receptor down-regulators may exhibit clinical efficacy in the treatment of gynecological malignancies due to their pure estrogen receptor antagonist properties. However, up to now data are still limited and some unsolved questions remain. Fulvestrant has poor oral bioavailability and low pharmacodynamic characteristics. Further trials are required to examine new selective estrogen receptor down-regulator agents with better pharmacodynamic and pharmacokinetic profiles.
Collapse
Affiliation(s)
- Stefano Bogliolo
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Chiara Cassani
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Mattia Dominoni
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Anna Orlandini
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Simone Ferrero
- b Department of Obstetrics and Gynaecology , IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, University of Genoa , Genoa , Italy
| | - Anna Daniela Iacobone
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Franco Viazzo
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Pier Luigi Venturini
- b Department of Obstetrics and Gynaecology , IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, University of Genoa , Genoa , Italy
| | - Arsenio Spinillo
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Barbara Gardella
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| |
Collapse
|
18
|
Tecalco-Cruz AC, Ramírez-Jarquín JO. Mechanisms that Increase Stability of Estrogen Receptor Alpha in Breast Cancer. Clin Breast Cancer 2016; 17:1-10. [PMID: 27561704 DOI: 10.1016/j.clbc.2016.07.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/29/2016] [Accepted: 07/20/2016] [Indexed: 12/20/2022]
Abstract
Estrogen receptor alpha (ER) is a transcriptional regulator that controls the expression of genes related to cellular proliferation and differentiation in normal mammary tissue. However, the expression, abundance, and activity of this receptor are increased in 70% of breast cancers. The ER upregulation is facilitated by several molecular mechanisms, including protein stability, which represents an important strategy to maintain an active and functional repertoire of ER. Several proteins interact and protect ER from degradation by the ubiquitin-proteasome system. Through diverse mechanisms, these proteins prevent polyubiquitination and degradation of ER, leading to an increase in ER protein levels; consequently, estrogen signaling and its physiologic effects are enhanced in breast cancer cells. Thus, increased protein stability seems to be one of the main reasons that ER is upregulated in breast cancer. Here, we highlight findings on the proteins and mechanisms that participate directly or indirectly in ER stability and their relevance to breast cancer.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F., Mexico.
| | - Josué O Ramírez-Jarquín
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| |
Collapse
|
19
|
Leclercq G, Laïos I, Elie-Caille C, Leiber D, Laurent G, Lesniewska E, Tanfin Z, Jacquot Y. ERα dimerization: a key factor for the weak estrogenic activity of an ERα modulator unable to compete with estradiol in binding assays. J Recept Signal Transduct Res 2016; 37:149-166. [PMID: 27400858 DOI: 10.1080/10799893.2016.1203940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Estrothiazine (ESTZ) is a weak estrogen sharing structural similarities with coumestrol. ESTZ failed to compete with [3H]17β-estradiol ([3H]17β-E2) for binding to the estrogen receptor α (ERα), questioning its ability to interact with the receptor. However, detection by atomic force spectroscopy (AFS) of an ESTZ-induced ERα dimerization has eliminated any remaining doubts. The effect of the compound on the proliferation of ERα-positive and negative breast cancer cells confirmed the requirement of the receptor. The efficiency of ESTZ in MCF-7 cells was weak without any potency to modify the proliferation profile of estradiol and coumestrol. Growth enhancement was associated with a proteasomal degradation of ERα without substantial recruitment of LxxLL coactivators. This may be related to an unusual delay between the acquisition by the receptor of an ERE-binding capacity and the subsequent estrogen-dependent transcription. A complementary ability to enhance TPA-induced AP-1 transcription was observed, even at concentrations insufficient to activate the ERα, suggesting a partly independent mechanism. ESTZ also rapidly and transiently activated ERK1/2 likely through membrane estrogenic pathways provoking a reorganization of the actin network. Finally, the systematic absence of biological responses with an ESTZ derivative unable to induce ERα dimerization stresses the importance of this step in the action of the compound, as reported for conventional estrogens. In view of the existence of many other ERα modulators (endocrine disruptors such as, for example, pesticides, environmental contaminants or phytoestrogens) with extremely weak or similar apparent lack of binding ability, our work may appear as pilot investigation for assessing their mechanism of action.
Collapse
Affiliation(s)
- Guy Leclercq
- a Laboratoire J.-C. Heuson de Cancérologie Mammaire , Université Libre de Bruxelles (U.L.B.), Institut Jules Bordet , Brussels , Belgium
| | - Ioanna Laïos
- a Laboratoire J.-C. Heuson de Cancérologie Mammaire , Université Libre de Bruxelles (U.L.B.), Institut Jules Bordet , Brussels , Belgium
| | - Céline Elie-Caille
- b Institut FEMTO-ST, CNRS UMR 6174, Université de Bourgogne Franche-Comté , Besançon , France
| | - Denis Leiber
- c Laboratoire Signalisation et Régulations Cellulaires , Institut de Biochimie et de Biologie Moléculaire et Cellulaire, CNRS UMR 8619, Université Paris-Sud , Orsay Cedex , France.,d INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Université d'Angers , Angers , France
| | - Guy Laurent
- e Service d'Histologie et de Cytologie Expérimentale, Faculté de Médecine et de Pharmacie , Université de Mons-Hainaut , Mons , Belgium
| | - Eric Lesniewska
- f ICB, CNRS UMR 6303, Université de Bourgogne Franche-Comté , Dijon , France
| | - Zahra Tanfin
- c Laboratoire Signalisation et Régulations Cellulaires , Institut de Biochimie et de Biologie Moléculaire et Cellulaire, CNRS UMR 8619, Université Paris-Sud , Orsay Cedex , France
| | - Yves Jacquot
- g Département de Chimie, CNRS UMR 7203 LBM , Sorbonne Universités - UPMC Univ Paris 06, Ecole Normale Supérieure, PSL Research University , Paris , France
| |
Collapse
|
20
|
Amita M, Takahashi T, Igarashi H, Nagase S. Clomiphene citrate down-regulates estrogen receptor-α through the ubiquitin-proteasome pathway in a human endometrial cancer cell line. Mol Cell Endocrinol 2016; 428:142-7. [PMID: 27033325 DOI: 10.1016/j.mce.2016.03.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 02/07/2023]
Abstract
We examined how clomiphene citrate (CC) reduces estrogen receptor-α (ERα) in a human endometrial cancer cell line. Ishikawa human endometrial cancer cells were treated with ERα ligands such as 17β-estradiol (E2), CC, and the pure antiestrogen, ICI 182,780 (ICI). Thereafter, the expression levels of ERα protein and mRNA were analyzed by western blot and real-time quantitative PCR, respectively, and those of ubiquitinated ERα were analyzed by immunoprecipitation of ERα followed by immunoblotting with an anti-ubiquitin antibody. The expression levels of ERα protein after treatment with E2, CC, and ICI were significantly decreased compared to pre-treatment levels without a corresponding decrease in ERα mRNA. These ligands significantly increased the levels of ubiquitinated ERα compared to vehicle treatment. Co-treatment with the proteasome inhibitor, MG132, abrogated the decrease in ERα levels caused by treatment with the ligands only. We demonstrated, for the first time, a CC-induced decrease in ERα mediated by the ubiquitin-proteasome pathway in human endometrial cancer cells.
Collapse
Affiliation(s)
- Mitsuyoshi Amita
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| | - Toshifumi Takahashi
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan.
| | - Hideki Igarashi
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| |
Collapse
|
21
|
Boisen MM, Andersen CL, Sreekumar S, Stern AM, Oesterreich S. Treating gynecologic malignancies with selective estrogen receptor downregulators (SERDs): promise and challenges. Mol Cell Endocrinol 2015; 418 Pt 3:322-33. [PMID: 26276546 DOI: 10.1016/j.mce.2015.04.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 04/16/2015] [Accepted: 04/16/2015] [Indexed: 02/07/2023]
Abstract
Endometrial and ovarian cancers are estrogen-dependent gynecologic malignancies. Although many are estrogen receptor (ER) positive, treatment with the selective estrogen receptor modulator (SERM) tamoxifen, a tissue selective partial-agonist, has demonstrated only modest clinical benefit. Selective estrogen receptor downregulators (SERDs) are pure ER antagonists showing a benefit for advanced ER positive breast cancer, which has bolstered their potential use for ER positive gynecologic malignancies. We summarize these preclinical and clinical data, suggesting that a subpopulation of patients with endometrial or ovarian cancer exists in which treatment with SERDs results in improved outcome. However, the full potential of SERDs for a gynecologic malignancies will be realized only when the appropriate predictive biomarkers are identified. Additionally, a further understanding ER signaling in the context of ovarian and endometrial tissues that appear to involve c-Src and other kinase pathways is needed to successfully address the emergence of resistance with rationally designed combination therapies.
Collapse
Affiliation(s)
- Michelle M Boisen
- Division of Gynecologic Oncology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | - Courtney L Andersen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine Molecular Pharmacology Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sreeja Sreekumar
- Women's Cancer Research Center, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute and the Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steffi Oesterreich
- University of Pittsburgh Cancer Institute, Department of Pharmacology and Chemical Biology, Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Casa AJ, Hochbaum D, Sreekumar S, Oesterreich S, Lee AV. The estrogen receptor alpha nuclear localization sequence is critical for fulvestrant-induced degradation of the receptor. Mol Cell Endocrinol 2015; 415:76-86. [PMID: 26272024 DOI: 10.1016/j.mce.2015.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/05/2015] [Accepted: 08/05/2015] [Indexed: 02/07/2023]
Abstract
Fulvestrant, a selective estrogen receptor down-regulator (SERD) is a pure competitive antagonist of estrogen receptor alpha (ERα). Fulvestrant binds ERα and reduces the receptor's half-life by increasing protein turnover, however, its mechanism of action is not fully understood. In this study, we show that removal of the ERα nuclear localization sequence (ERΔNLS) resulted in a predominantly cytoplasmic ERα that was degraded in response to 17-β-estradiol (E2) but was resistant to degradation by fulvestrant. ERΔNLS bound the ligands and exhibited receptor interaction similar to ERα, indicating that the lack of degradation was not due to disruption of these processes. Forcing ERΔNLS into the nucleus with a heterologous SV40-NLS did not restore degradation, suggesting that the NLS domain itself, and not merely receptor localization, is critical for fulvestrant-induced ERα degradation. Indeed, cloning of the endogenous ERα NLS onto the N-terminus of ERΔNLS significantly restored both its nuclear localization and turnover in response to fulvestrant. Moreover, mutation of the sumoylation targets K266 and K268 within the NLS impaired fulvestrant-induced ERα degradation. In conclusion, our study provides evidence for the unique role of the ERα NLS in fulvestrant-induced degradation of the receptor.
Collapse
Affiliation(s)
- Angelo J Casa
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniel Hochbaum
- Women's Cancer Research Center, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | - Sreeja Sreekumar
- Women's Cancer Research Center, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | - Adrian V Lee
- Women's Cancer Research Center, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| |
Collapse
|
23
|
Lai A, Kahraman M, Govek S, Nagasawa J, Bonnefous C, Julien J, Douglas K, Sensintaffar J, Lu N, Lee KJ, Aparicio A, Kaufman J, Qian J, Shao G, Prudente R, Moon MJ, Joseph JD, Darimont B, Brigham D, Grillot K, Heyman R, Rix PJ, Hager JH, Smith ND. Identification of GDC-0810 (ARN-810), an Orally Bioavailable Selective Estrogen Receptor Degrader (SERD) that Demonstrates Robust Activity in Tamoxifen-Resistant Breast Cancer Xenografts. J Med Chem 2015; 58:4888-904. [PMID: 25879485 DOI: 10.1021/acs.jmedchem.5b00054] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Approximately 80% of breast cancers are estrogen receptor alpha (ER-α) positive, and although women typically initially respond well to antihormonal therapies such as tamoxifen and aromatase inhibitors, resistance often emerges. Although a variety of resistance mechanism may be at play in this state, there is evidence that in many cases the ER still plays a central role, including mutations in the ER leading to constitutively active receptor. Fulvestrant is a steroid-based, selective estrogen receptor degrader (SERD) that both antagonizes and degrades ER-α and is active in patients who have progressed on antihormonal agents. However, fulvestrant suffers from poor pharmaceutical properties and must be administered by intramuscular injections that limit the total amount of drug that can be administered and hence lead to the potential for incomplete receptor blockade. We describe the identification and characterization of a series of small-molecule, orally bioavailable SERDs which are potent antagonists and degraders of ER-α and in which the ER-α degrading properties were prospectively optimized. The lead compound 11l (GDC-0810 or ARN-810) demonstrates robust activity in models of tamoxifen-sensitive and tamoxifen-resistant breast cancer, and is currently in clinical trials in women with locally advanced or metastatic estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Andiliy Lai
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Mehmet Kahraman
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Steven Govek
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Johnny Nagasawa
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Celine Bonnefous
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Jackie Julien
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Karensa Douglas
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - John Sensintaffar
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Nhin Lu
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Kyoung-Jin Lee
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Anna Aparicio
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Josh Kaufman
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Jing Qian
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Gang Shao
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Rene Prudente
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Michael J Moon
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - James D Joseph
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Beatrice Darimont
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Daniel Brigham
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Kate Grillot
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Richard Heyman
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Peter J Rix
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Jeffrey H Hager
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| | - Nicholas D Smith
- †Department of Chemistry, ‡Department of Biology, §Department of Drug Safety and Disposition, Seragon Pharmaceuticals, 12780 El Camino Real, Suite 302, San Diego, California 92130, United States
| |
Collapse
|
24
|
Urbinati G, Marsaud V, Nicolas V, Vergnaud-Gauduchon J, Renoir JM. Liposomal trichostatin A: therapeutic potential in hormone-dependent and -independent breast cancer xenograft models. Horm Mol Biol Clin Investig 2015; 6:215-25. [PMID: 25961258 DOI: 10.1515/hmbci.2011.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 01/12/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND Trichostatin A (TSA) is one of the most potent histone deacetylase inhibitors (HDACi) in vitro but it lacks biological activity in vivo when injected intravenously owing to its fast metabolism. MATERIALS AND METHODS TSA was incorporated into Stealth® liposomes (TSA-lipo) at a high loading and its anticancer activity was evaluated in several types of breast cancer cells and xenografts. RESULTS In estrogen receptor α (ERα)-positive MCF-7 and T47-D cells, TSA induced a long-term degradation of cyclin A and a proteasome-dependent loss of ERα and cyclin D1, allowed derepression of p21WAF1/CIP1, HDAC1 and RhoB GTPase, concomitantly with blockade in G2/M of the cell cycle and apoptosis induction. In MDA-MB-231 (MDA) and SKBr-3 cells, TSA increased ERα mRNA and p21WAF1/CIP1 protein expression, but decreased cyclin A with a G2/M blockade and cleavage of polyADP-ribose polymerase (PARP). No significant restoration of any ER protein was noticed in any cells. TSA-lipo markedly inhibited tumor growth in MCF-7 and MDA cells xenografts following intravenous injection. Their anticancer effects were characterized by inhibition of Ki-67 labeling, the inhibition of tumor vasculature and an increase of p21WAF1/CIP1 in both tumors. In MCF-7 cell tumors, enhanced RhoB accumulation in the cytoplasm of epithelial cells was noticed, inversely to ERα that was strongly decreased. CONCLUSION Such anticancer activity of TSA-lipo is exp-lained by the protection provided by HDACi encapsulation and by the strong tumor accumulation of the nanocarriers as revealed by fluorescence confocal microscopy experi-ments. Together with its lack of toxicity, the enhanced stability of TSA-lipo in vivo justifies its development for therapeutic use in the treatment estradiol-dependent and -independent breast cancers.
Collapse
|
25
|
Callis R, Rabow A, Tonge M, Bradbury R, Challinor M, Roberts K, Jones K, Walker G. A Screening Assay Cascade to Identify and Characterize Novel Selective Estrogen Receptor Downregulators (SERDs). ACTA ACUST UNITED AC 2015; 20:748-59. [DOI: 10.1177/1087057115580298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/27/2015] [Indexed: 11/16/2022]
Abstract
Here, we describe an approach to identify novel selective estrogen receptor downregulator (SERD) compounds with improved properties such as oral bioavailability and the potential of increased efficacy compared to currently marketed drug treatments. Previously, methodologies such as Western blotting and transient cell reporter assays have been used to identify and characterize SERD compounds, but such approaches can be limited due to low throughput and sensitivity, respectively. We have used an endogenous cell-imaging strategy that has both the throughput and sensitivity to support a large-scale hit-to-lead program to identify novel compounds. A screening cascade with a suite of assays has been developed to characterize compounds that modulate estrogen receptor α (ERα)-mediated signaling or downregulate ERα levels in cells. Initially, from a focused high-throughput screening, novel ERα binders were identified that could be modified chemically into ERα downregulators. Following this, cellular assays helped determine the mechanism of action of compounds to distinguish between on-target and off-target compounds and differentiate SERDs, selective estrogen receptor modulator (SERM) compounds, and agonist ERα ligands. Data are shown to exemplify the characterization of ERα-mediated signaling inhibitors using a selection of literature compounds and illustrate how this cascade has been used to drive the chemical design of novel SERD compounds.
Collapse
Affiliation(s)
- Rowena Callis
- Discovery Sciences, AstraZeneca, Macclesfield, Cheshire, UK
| | - Alfred Rabow
- Oncology Innovative Medicines Unit, AstraZeneca, Macclesfield, Cheshire, UK
| | - Michael Tonge
- Discovery Sciences, AstraZeneca, Macclesfield, Cheshire, UK
| | - Robert Bradbury
- Oncology Innovative Medicines Unit, AstraZeneca, Macclesfield, Cheshire, UK
| | | | - Karen Roberts
- Discovery Sciences, AstraZeneca, Macclesfield, Cheshire, UK
| | - Karen Jones
- Discovery Sciences, AstraZeneca, Macclesfield, Cheshire, UK
| | - Graeme Walker
- Discovery Sciences, AstraZeneca, Macclesfield, Cheshire, UK
| |
Collapse
|
26
|
Kundu P, Li M, Lu R, Stefani E, Toro L. Regulation of transcriptional activation function of rat estrogen receptor α (ERα) by novel C-terminal splice inserts. Mol Cell Endocrinol 2015; 401:202-12. [PMID: 25451981 PMCID: PMC4312711 DOI: 10.1016/j.mce.2014.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 11/01/2014] [Accepted: 11/03/2014] [Indexed: 01/19/2023]
Abstract
Estrogen receptor α (ERα) mediates estrogen diverse actions on tissues. ERα gene has eight constitutively expressing exons and is known to have multiple isoforms generated by alternative initiation of transcription and splicing events including exon skipping. We have discovered two novel exons inserted between exon 5 and 6 of rat ERα that can add independently or in tandem 18 and 14 amino acids to the estrogen binding/activator function 2 domain of the receptor. Their transcript expression is three to six fold higher in heart compared to brain, aorta, liver, ovary and uterus. In heart, the new variants increased ~2 fold with animal growth from prenatal to adulthood, and had a minor increment in aged animals (28 months). Inclusion of these exons yields a receptor with practically no binding capacity for estrogen and reduced dimerization. The new variants show nuclear localization but are less efficient in binding to estrogen responsive elements (EREs) and failed to transcriptionally activate promoters containing EREs (mSlo, KCNE2). Thus, the new variants can regulate the wild-type receptor function and may contribute to the regulatory action of estrogen, especially in the maturing heart where they are more abundant.
Collapse
Affiliation(s)
- Pallob Kundu
- Department of Anesthesiology, University of California, Los Angeles, CA 90095, USA; Division of Plant Biology, Bose Institute, Kolkata 700054, India.
| | - Min Li
- Department of Anesthesiology, University of California, Los Angeles, CA 90095, USA
| | - Rong Lu
- Department of Anesthesiology, University of California, Los Angeles, CA 90095, USA
| | - Enrico Stefani
- Department of Anesthesiology, University of California, Los Angeles, CA 90095, USA; Department of Physiology, University of California, Los Angeles, CA 90095, USA; Department of Brain Research Institute, University of California, Los Angeles, CA 90095, USA; Department of Cardiovascular Research Laboratory, University of California, Los Angeles, CA 90095, USA
| | - Ligia Toro
- Department of Anesthesiology, University of California, Los Angeles, CA 90095, USA; Department of Brain Research Institute, University of California, Los Angeles, CA 90095, USA; Department of Cardiovascular Research Laboratory, University of California, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
27
|
Tian D, Solodin NM, Rajbhandari P, Bjorklund K, Alarid ET, Kreeger PK. A kinetic model identifies phosphorylated estrogen receptor-α (ERα) as a critical regulator of ERα dynamics in breast cancer. FASEB J 2015; 29:2022-31. [PMID: 25648997 DOI: 10.1096/fj.14-265637] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/05/2015] [Indexed: 11/11/2022]
Abstract
Receptor levels are a key mechanism by which cells regulate their response to stimuli. The levels of estrogen receptor-α (ERα) impact breast cancer cell proliferation and are used to predict prognosis and sensitivity to endocrine therapy. Despite the clinical application of this information, it remains unclear how different cellular processes interact as a system to control ERα levels. To address this question, experimental results from the ERα-positive human breast cancer cell line (MCF-7) treated with 17-β-estradiol or vehicle control were used to develop a mass-action kinetic model of ERα regulation. Model analysis determined that RNA dynamics could be captured through phosphorylated ERα (pERα)-dependent feedback on transcription. Experimental analysis confirmed that pERα-S118 binds to the estrogen receptor-1 (ESR1) promoter, suggesting that pERα can feedback on ESR1 transcription. Protein dynamics required a separate mechanism in which the degradation rate for pERα was 8.3-fold higher than nonphosphorylated ERα. Using a model with both mechanisms, the root mean square error was 0.078. Sensitivity analysis of this combined model determined that while multiple mechanisms regulate ERα levels, pERα-dependent feedback elicited the strongest effect. Combined, our computational and experimental results identify phosphorylation of ERα as a critical decision point that coordinates the cellular circuitry to regulate ERα levels.
Collapse
Affiliation(s)
- Dan Tian
- *Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Natalia M Solodin
- *Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Prashant Rajbhandari
- *Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Kelsi Bjorklund
- *Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Elaine T Alarid
- *Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Pamela K Kreeger
- *Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; and University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| |
Collapse
|
28
|
Xiao L, Chang AK, Zang MX, Bi H, Li S, Wang M, Xing X, Wu H. Induction of the CLOCK gene by E2-ERα signaling promotes the proliferation of breast cancer cells. PLoS One 2014; 9:e95878. [PMID: 24789043 PMCID: PMC4008427 DOI: 10.1371/journal.pone.0095878] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 04/01/2014] [Indexed: 11/19/2022] Open
Abstract
Growing genetic and epidemiological evidence suggests a direct connection between the disruption of circadian rhythm and breast cancer. Moreover, the expression of several molecular components constituting the circadian clock machinery has been found to be modulated by estrogen-estrogen receptor α (E2-ERα) signaling in ERα-positive breast cancer cells. In this study, we investigated the regulation of CLOCK expression by ERα and its roles in cell proliferation. Immunohistochemical analysis of human breast tumor samples revealed high expression of CLOCK in ERα-positive breast tumor samples. Subsequent experiments using ERα-positive human breast cancer cell lines showed that both protein and mRNA levels of CLOCK were up-regulated by E2 and ERα. In these cells, E2 promoted the binding of ERα to the EREs (estrogen-response elements) of CLOCK promoter, thereby up-regulating the transcription of CLOCK. Knockdown of CLOCK attenuated cell proliferation in ERα-positive breast cancer cells. Taken together, these results demonstrated that CLOCK could be an important gene that mediates cell proliferation in breast cancer cells.
Collapse
Affiliation(s)
- Liyun Xiao
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Alan K. Chang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Ming-Xi Zang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hailian Bi
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Shujing Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Miao Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Xinrong Xing
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Huijian Wu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
- School of Life Science and Medicine, Dalian University of Technology, Panjin, China
- * E-mail:
| |
Collapse
|
29
|
Pereira MFN, Fernandes SAF, Nascimento AR, Siu ER, Hess RA, Oliveira CA, Porto CS, Lazari MFM. Effects of the oestrogen receptor antagonist Fulvestrant on expression of genes that affect organization of the epididymal epithelium. Andrology 2014; 2:559-71. [PMID: 24782439 DOI: 10.1111/j.2047-2927.2014.00219.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/18/2014] [Accepted: 03/21/2014] [Indexed: 12/20/2022]
Abstract
The role of oestrogens in epididymal function is still unclear. Knockout of the oestrogen receptor ESR1 (Esr1(-/-) ) or treatment with the anti-oestrogen Fulvestrant affect epididymal milieu and sperm motility. We investigated the effect of in vivo treatment of rats with Fulvestrant on: (i) expression of genes that may be important for the architecture and function of the epididymal epithelium: prominins 1 and 2, metalloproteinase 7, claudin 7, beta-catenin and cadherin 13, and (ii) levels of oestradiol and testosterone, and expression of oestrogen and androgen receptors, in the initial segment (IS), caput, corpus and cauda epididymis. Fulvestrant (i) reduced gene expression of prominin 1 (variant 1) in the caput, reduced prominin 1 protein content in the caput epididymis and in the efferent ductules, and increased the localization of prominin 1 in microvilli of the caput and corpus; (ii) reduced gene expression of prominin 2 in the corpus and cauda epididymis; (iii) increased the metalloproteinase 7 content in the apical region of principal cells from IS/caput; (iv) reduced in the corpus epididymis, but increased in the efferent ductules, the cadherin 13 mRNA level; (v) reduced testosterone but increased oestradiol levels in the corpus and cauda; (vi) increased the androgen receptor protein content in all regions of the epididymis, and the oestrogen receptor GPER in the corpus and cauda epididymis. In conclusion, treatment with Fulvestrant induced regional-specific changes in hormonal and steroid receptor content, and affected expression of proteins important for epithelial organization and absorption/secretion. The mechanisms of oestrogen action may differ among epididymal regions, which may contribute to determine region-specific sperm functions.
Collapse
Affiliation(s)
- M F N Pereira
- Section of Experimental Endocrinology, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Regulation of estrogen receptor signaling in breast carcinogenesis and breast cancer therapy. Cell Mol Life Sci 2014; 71:1549. [PMID: 25031550 PMCID: PMC3962223 DOI: 10.1007/s00018-013-1376-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 12/19/2022]
Abstract
Estrogen and estrogen receptors (ERs) are critical regulators of breast epithelial cell proliferation, differentiation, and apoptosis. Compromised signaling vis-à-vis the estrogen receptor is believed to be a major contributing factor in the malignancy of breast cells. Targeting the ER signaling pathway has been a focal point in the development of breast cancer therapy. Although approximately 75 % of breast cancer patients are classified as luminal type (ER(+)), which predicts for response to endocrine-based therapy; however, innate or acquired resistance to endocrine-based drugs remains a serious challenge. The complexity of regulation for estrogen signaling coupled with the crosstalk of other oncogenic signaling pathways is a reason for endocrine therapy resistance. Alternative strategies that target novel molecular mechanisms are necessary to overcome this current and urgent gap in therapy. A thorough analysis of estrogen-signaling regulation is critical. In this review article, we will summarize current insights into the regulation of estrogen signaling as related to breast carcinogenesis and breast cancer therapy.
Collapse
|
31
|
Estrogen and pure antiestrogen fulvestrant (ICI 182 780) augment cell-matrigel adhesion of MCF-7 breast cancer cells through a novel G protein coupled estrogen receptor (GPR30)-to-calpain signaling axis. Toxicol Appl Pharmacol 2014; 275:176-81. [PMID: 24440569 DOI: 10.1016/j.taap.2014.01.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/08/2014] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
Fulvestrant (ICI 182 780, ICI) has been used in treating patients with hormone-sensitive breast cancer, yet initial or acquired resistance to endocrine therapies frequently arises and, in particular, cancer recurs as metastasis. We demonstrate here that both 17-beta-estradiol (E2) and ICI enhance cell adhesion to matrigel in MCF-7 breast cancer cells, with increased autolysis of calpain 1 (large subunit) and proteolysis of focal adhesion kinase (FAK), indicating calpain activation. Additionally, either E2 or ICI induced down-regulation of estrogen receptor α without affecting G protein coupled estrogen receptor 30 (GPR30) expression. Interestingly, GPR30 agonist G1 triggered calpain 1 autolysis but not calpain 2, whereas ER agonist diethylstilbestrol caused no apparent calpain autolysis. Furthermore, the actions of E2 and ICI on calpain and cell adhesion were tremendously suppressed by G15, or knockdown of GPR30. E2 and ICI also induced phosphorylation of extracellular regulated protein kinases 1 and 2 (ERK1/2), and suppression of ERK1/2 phosphorylation by U0126 profoundly impeded calpain activation triggered by estrogenic and antiestrogenic stimulations indicating implication of ERK1/2 in the GPR30-mediated action. Lastly, the E2- or ICI-induced cell adhesion was dramatically impaired by calpain-specific inhibitors, ALLN or calpeptin, suggesting requirement of calpain in the GPR30-associated action. These data show that enhanced cell adhesion by E2 and ICI occurs via a novel GPR30-ERK1/2-calpain pathway. Our results indicate that targeting the GPR30 signaling may be a potential strategy to reduce metastasis and improve the efficacy of antiestrogens in treatment of advanced breast cancer.
Collapse
|
32
|
Yom CK, Lee KM, Han W, Kim SW, Kim HS, Moon BI, Jeong KY, Im SA, Noh DY. Leptin as a potential target for estrogen receptor-positive breast cancer. J Breast Cancer 2013; 16:138-45. [PMID: 23843844 PMCID: PMC3706857 DOI: 10.4048/jbc.2013.16.2.138] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 04/02/2013] [Indexed: 11/30/2022] Open
Abstract
Purpose Leptin is a potent adipokine that plays a significant role in tumor development and the progression of breast cancer. The aim of this study was to evaluate whether leptin affects the response to tamoxifen treatment in estrogen receptor (ER)-positive breast cancer cells. Methods Leptin, leptin receptor (Ob-R), and activation of signaling pathways were studied by Western immunoblotting. The effects of leptin on tamoxifen-dependent growth inhibition were studied in MCF-7 and T-47D cells. Results Leptin was expressed in MCF-7 and T-47D and had a proliferative effect on MCF-7 cells. Leptin significantly inhibited the antiestrogenic effect of tamoxifen in both cells only under β-estradiol (E2) (20 nM) conditions. In MCF-7, the inhibitory effect against tamoxifen was a result from the activation of the ERK1/2 and STAT3 signal transduction pathway. Conclusion Leptin interferes with the effects of tamoxifen under E2 stimulated conditions in ER-positive breast cancer cells. These results imply that inhibition of leptin is expected to enhance the response to tamoxifen in ER-positive breast cancer cells, and, therefore, could be a promising way to overcome endocrine resistance.
Collapse
Affiliation(s)
- Cha Kyong Yom
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yeh WL, Shioda K, Coser KR, Rivizzigno D, McSweeney KR, Shioda T. Fulvestrant-induced cell death and proteasomal degradation of estrogen receptor α protein in MCF-7 cells require the CSK c-Src tyrosine kinase. PLoS One 2013; 8:e60889. [PMID: 23593342 PMCID: PMC3617152 DOI: 10.1371/journal.pone.0060889] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 03/06/2013] [Indexed: 12/19/2022] Open
Abstract
Fulvestrant is a representative pure antiestrogen and a Selective Estrogen Receptor Down-regulator (SERD). In contrast to the Selective Estrogen Receptor Modulators (SERMs) such as 4-hydroxytamoxifen that bind to estrogen receptor α (ERα) as antagonists or partial agonists, fulvestrant causes proteasomal degradation of ERα protein, shutting down the estrogen signaling to induce proliferation arrest and apoptosis of estrogen-dependent breast cancer cells. We performed genome-wide RNAi knockdown screenings for protein kinases required for fulvestrant-induced apoptosis of the MCF-7 estrogen-dependent human breast caner cells and identified the c-Src tyrosine kinase (CSK), a negative regulator of the oncoprotein c-Src and related protein tyrosine kinases, as one of the necessary molecules. Whereas RNAi knockdown of CSK in MCF-7 cells by shRNA-expressing lentiviruses strongly suppressed fulvestrant-induced cell death, CSK knockdown did not affect cytocidal actions of 4-hydroxytamoxifen or paclitaxel, a chemotherapeutic agent. In the absence of CSK, fulvestrant-induced proteasomal degradation of ERα protein was suppressed in both MCF-7 and T47D estrogen-dependent breast cancer cells whereas the TP53-mutated T47D cells were resistant to the cytocidal action of fulvestrant in the presence or absence of CSK. MCF-7 cell sensitivities to fulvestrant-induced cell death or ERα protein degradation was not affected by small-molecular-weight inhibitors of the tyrosine kinase activity of c-Src, suggesting possible involvement of other signaling molecules in CSK-dependent MCF-7 cell death induced by fulvestrant. Our observations suggest the importance of CSK in the determination of cellular sensitivity to the cytocidal action of fulvestrant.
Collapse
Affiliation(s)
- Wei-Lan Yeh
- Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Keiko Shioda
- Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Kathryn R. Coser
- Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Danielle Rivizzigno
- Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Kristen R. McSweeney
- Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Toshi Shioda
- Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
34
|
Rajbhandari P, Schalper KA, Solodin NM, Ellison-Zelski SJ, Ping Lu K, Rimm DL, Alarid ET. Pin1 modulates ERα levels in breast cancer through inhibition of phosphorylation-dependent ubiquitination and degradation. Oncogene 2013; 33:1438-47. [PMID: 23542176 DOI: 10.1038/onc.2013.78] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 01/28/2013] [Accepted: 02/01/2013] [Indexed: 12/20/2022]
Abstract
Estrogen receptor-alpha (ERα) is an important biomarker used to classify and direct therapy decisions in breast cancer (BC). Both ERα protein and its transcript, ESR1, are used to predict response to tamoxifen therapy, yet certain tumors have discordant levels of ERα protein and ESR1, which is currently unexplained. Cellular ERα protein levels can be controlled post-translationally by the ubiquitin-proteasome pathway through a mechanism that depends on phosphorylation at residue S118. Phospho-S118 (pS118-ERα) is a substrate for the peptidyl prolyl isomerase, Pin1, which mediates cis-trans isomerization of the pS118-P119 bond to enhance ERα transcriptional function. Here, we demonstrate that Pin1 can increase ERα protein without affecting ESR1 transcript levels by inhibiting proteasome-dependent receptor degradation. Pin1 disrupts ERα ubiquitination by interfering with receptor interactions with the E3 ligase, E6AP, which also is shown to bind pS118-ERα. Quantitative in situ assessments of ERα protein, ESR1, and Pin1 in human tumors from a retrospective cohort show that Pin1 levels correlate with ERα protein but not to ESR1 levels. These data show that ERα protein is post-translationally regulated by Pin1 in a proportion of breast carcinomas. As Pin1 impacts both ERα protein levels and transactivation function, these data implicate Pin1 as a potential surrogate marker for predicting outcome of ERα-positive BC.
Collapse
Affiliation(s)
- P Rajbhandari
- Department of Oncology, UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - K A Schalper
- Department of Pathology, Yale University Medical School, New Haven, CT, USA
| | - N M Solodin
- Department of Oncology, UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - S J Ellison-Zelski
- Department of Oncology, UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - K Ping Lu
- Department of Medicine, Cancer Biology Program, Beth Isreal Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - D L Rimm
- Department of Pathology, Yale University Medical School, New Haven, CT, USA
| | - E T Alarid
- Department of Oncology, UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
35
|
Tabatadze N, Smejkalova T, Woolley CS. Distribution and posttranslational modification of synaptic ERα in the adult female rat hippocampus. Endocrinology 2013; 154. [PMID: 23183182 PMCID: PMC3548183 DOI: 10.1210/en.2012-1870] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Acute 17β-estradiol (E2) signaling in the brain is mediated by extranuclear estrogen receptors. Here we used biochemical methods to investigate the distribution, posttranslational modification, and E2 regulation of estrogen receptor-α (ERα) in synaptosomal fractions isolated by differential centrifugation from the adult female rat hippocampus. We find that ERα is concentrated presynaptically and is highly enriched with synaptic vesicles. Immunoisolation of vesicles using vesicle subtype-specific markers showed that ERα is associated with both glutamate and γ-aminobutyric acid-containing neurotransmitter vesicles as well as with some large dense core vesicles. Experiments using broad spectrum and residue-specific phosphatases indicated that a portion of ERα in synaptosomal fractions is phosphorylated at serine/threonine residues leading to a mobility shift in SDS-PAGE and creating a double band on Western blots. The phosphorylated form of ERα runs in the upper of the two bands and is particularly concentrated with synaptic vesicles. Finally, we used E2 with or without the acyl protein thioesterase 1 inhibitor, Palmostatin B, to show that 20 min of E2 treatment of hippocampal slices depletes ERα from the synaptosomal membrane by depalmitoylation. We found no evidence that E2 regulates phosphorylation of synaptosomal ERα on this time scale. These studies begin to fill the gap between detailed molecular characterization of extranuclear ERα in previous in vitro studies and acute E2 modulation of hippocampal synapses in the adult brain.
Collapse
Affiliation(s)
- Nino Tabatadze
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | | | | |
Collapse
|
36
|
Renoir JM, Marsaud V, Lazennec G. Estrogen receptor signaling as a target for novel breast cancer therapeutics. Biochem Pharmacol 2013; 85:449-65. [DOI: 10.1016/j.bcp.2012.10.018] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 10/11/2012] [Accepted: 10/19/2012] [Indexed: 02/07/2023]
|
37
|
Filis P, Kind PC, Spears N. Implantation failure in mice with a disruption in Phospholipase C beta 1 gene: lack of embryonic attachment, aberrant steroid hormone signalling and defective endocannabinoid metabolism. Mol Hum Reprod 2013; 19:290-301. [PMID: 23295235 DOI: 10.1093/molehr/gas067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Phospholipase C beta 1 (PLCβ1) is a downstream effector of G-protein-coupled receptor signalling and holds central roles in reproductive physiology. Mice with a disruption in the Plcβ1 gene are infertile with pleiotropic reproductive defects, the major reproductive block in females being implantation failure. Here, PLCβ1 was demonstrated at the luminal and glandular epithelia throughout the pre- and peri-implantation period, with transient stromal expression during 0.5-1.5 days post coitum (dpc). Examination of implantation sites at 4.5 dpc showed that in females lacking functional PLCβ1 (knock-out (KO) females), embryos failed to establish proper contact with the uterine epithelium. Proliferating luminal epithelial cells were evident in KO implantation sites, indicating failure to establish a receptive uterus. Real-time PCR demonstrated that KO implantation sites had aberrant ovarian steroid signalling, with high levels of estrogen receptor α, lactoferrin and amphiregulin mRNA, while immunohistochemistry revealed very low levels of estrogen receptor α protein, possibly due to rapid receptor turnover. KO implantation sites expressed markedly less fatty acid amide hydrolase and monoacylglycerol lipase, indicating that endocannabinoid metabolism was also affected. Collectively, our results show that PLCβ1 is essential for uterine preparation for implantation, and that defective PLCβ1-mediated signalling during implantation is associated with aberrant ovarian steroid signalling and endocannabinoid metabolism.
Collapse
Affiliation(s)
- Panayiotis Filis
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | | | | |
Collapse
|
38
|
Estrogen receptor prevents p53-dependent apoptosis in breast cancer. Proc Natl Acad Sci U S A 2012; 109:18060-5. [PMID: 23077249 DOI: 10.1073/pnas.1018858109] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
More than two-thirds of breast cancers express the estrogen receptor (ER) and depend on estrogen for growth and survival. Therapies targeting ER function, including aromatase inhibitors that block the production of estrogens and ER antagonists that alter ER transcriptional activity, play a central role in the treatment of ER+ breast cancers of all stages. In contrast to ER- breast cancers, which frequently harbor mutations in the p53 tumor suppressor, ER+ breast cancers are predominantly wild type for p53. Despite harboring wild-type p53, ER+ breast cancer cells are resistant to chemotherapy-induced apoptosis in the presence of estrogen. Using genome-wide approaches, we have addressed the mechanism by which ER antagonizes the proapoptotic function of p53. Interestingly, both ER agonists such as estradiol and the selective ER modulator (SERM) tamoxifen promote p53 antagonism. In contrast, the full ER antagonist fulvestrant blocks the ability of ER to inhibit p53-mediated cell death. This inhibition works through a mechanism involving the modulation of a subset of p53 and ER target genes that can predict the relapse-free survival of patients with ER+ breast cancer. These findings suggest an improved strategy for the treatment of ER+ breast cancer using antagonists that completely block ER action together with drugs that activate p53-mediated cell death.
Collapse
|
39
|
Renoir JM. Estradiol receptors in breast cancer cells: associated co-factors as targets for new therapeutic approaches. Steroids 2012; 77:1249-61. [PMID: 22917634 DOI: 10.1016/j.steroids.2012.07.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/18/2012] [Accepted: 07/25/2012] [Indexed: 02/07/2023]
Abstract
Estrogen receptors α (ERα) and β (ERβ) are nuclear receptors which transduce estradiol (E2) response in many tissues including the mammary gland and breast cancers (BC). They activate or inhibit specific genes involved in cell cycle progression and cell survival through multiple enzyme activities leading to malignant transformation. Hormone therapy (antiestrogens (AEs) and aromatase inhibitors (AIs) have been widely used to block the mitogenic action of E2 in patients with ER-positive BC. ERs act in concert with numerous other proteins outside and inside the nucleus where co-activators such as histone modifying enzymes help reaching optimum gene activation. Moreover, E2-mediated gene regulation can occur through ERs located at the plasma membrane or G protein-coupled estrogen receptor (GPER), triggering protein kinase signaling cascades. Classical AEs as well as AIs are inefficient to block the cascades of events emanating from the membrane and from E2 binding to GPER, leading patients to escape anti-hormone treatments and hormone therapy resistance. Many pathways are involved in resistance, mostly resulting from over-expression of growth factor membrane receptors, in particular the HER2/ErbB2 which can be inhibited by specific antibodies or tyrosine kinases inhibitors. Together with the Hsp90 molecular chaperone machinery, a complex interplay between ERs, co-activators, co-repressors and growth factor-activated membrane pathways represents potent targets which warrant to be manipulated alone and in combination to designing novel therapies. The discovery of new potential targets arising from micro array studies gives the opportunity to activate or inhibit different new ER-modulating effectors for innovative therapeutic interventions.
Collapse
|
40
|
Marino M, Pellegrini M, La Rosa P, Acconcia F. Susceptibility of estrogen receptor rapid responses to xenoestrogens: Physiological outcomes. Steroids 2012; 77:910-7. [PMID: 22410438 DOI: 10.1016/j.steroids.2012.02.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 02/13/2012] [Accepted: 02/24/2012] [Indexed: 02/06/2023]
Abstract
17β-Estradiol (E2) binding induces rapid modification in the conformation of its cognate receptors (i.e., ERα and ERβ). These allosteric changes allow the association of ERs with cell specific transcriptional cofactors, thus determining cellular contexts specific variations in gene expression. In addition, E2-ER complexes could also interact with membrane and cytosolic signal molecules triggering extra-nuclear signalling pathways. The synergy between these mechanisms is necessary for E2-induced pleiotropic actions in target tissues. Besides E2, the ER ligand binding domains can accommodate many other natural and synthetic ligands. Several of these compounds act as agonist or antagonist of ER transcriptional activity due to their ability to modify the interactions between ERs and transcriptional co-regulators. However, the ability of natural or manmade ER ligands to affect the extra-nuclear interactions of the ERs has been rarely evaluated. Here, the ability of two diet-derived flavonoids (i.e., naringenin and quercetin) and of the synthetic food-contaminant bisphenol A to modulate specifically ER extra-nuclear signalling pathways will be reported. All the tested compounds bind to both ER subtypes even if lesser than E2 activating divergent signal transduction pathways. In fact, in the presence of ERα, both naringenin and quercetin decouple ERα activities by specifically interfering with ERα membrane initiating signals. On the other hand, bisphenol A, but not flavonoids, maintains ERβ at the membrane thus impairing the activation of the downstream kinases. As a whole, extra-nuclear ER signals are highly susceptible to different ligands that, by unbalancing E2-induced cell functions drive cells to different functional endpoints.
Collapse
Affiliation(s)
- Maria Marino
- Department of Biology, University Roma TRE, viale G. Marconi, 446, I-00146 Rome, Italy.
| | | | | | | |
Collapse
|
41
|
Abstract
The selective estrogen receptor downregulator (SERD) fulvestrant can be used as second-line treatment for patients relapsing after treatment with tamoxifen, a selective estrogen receptor modulator (SERM). Unlike tamoxifen, SERDs are devoid of partial agonist activity. While the full antiestrogenicity of SERDs may result in part from their capacity to downregulate levels of estrogen receptor alpha (ERα) through proteasome-mediated degradation, SERDs are also fully antiestrogenic in the absence of increased receptor turnover in HepG2 cells. Here we report that SERDs induce the rapid and strong SUMOylation of ERα in ERα-positive and -negative cell lines, including HepG2 cells. Four sites of SUMOylation were identified by mass spectrometry analysis. In derivatives of the SERD ICI164,384, SUMOylation was dependent on the length of the side chain and correlated with full antiestrogenicity. Preventing SUMOylation by the overexpression of a SUMO-specific protease (SENP) deSUMOylase partially derepressed transcription in the presence of full antiestrogens in HepG2 cells without a corresponding increase in activity in the presence of agonists or of the SERM tamoxifen. Mutations increasing transcriptional activity in the presence of full antiestrogens reduced SUMOylation levels and suppressed stimulation by SENP1. Our results indicate that ERα SUMOylation contributes to full antiestrogenicity in the absence of accelerated receptor turnover.
Collapse
|
42
|
La Rosa P, Pesiri V, Leclercq G, Marino M, Acconcia F. Palmitoylation regulates 17β-estradiol-induced estrogen receptor-α degradation and transcriptional activity. Mol Endocrinol 2012; 26:762-74. [PMID: 22446104 DOI: 10.1210/me.2011-1208] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The estrogen receptor-α (ERα) is a transcription factor that regulates gene expression through the binding to its cognate hormone 17β-estradiol (E2). ERα transcriptional activity is regulated by E2-evoked 26S proteasome-mediated ERα degradation and ERα serine (S) residue 118 phosphorylation. Furthermore, ERα mediates fast cell responses to E2 through the activation of signaling cascades such as the MAPK/ERK and phosphoinositide-3-kinase/v-akt murine thymoma viral oncogene homolog 1 pathways. These E2 rapid effects require a population of the ERα located at the cell plasma membrane through palmitoylation, a dynamic enzymatic modification mediated by palmitoyl-acyl-transferases. However, whether membrane-initiated and transcriptional ERα activities integrate in a unique picture or represent parallel pathways still remains to be firmly clarified. Hence, we evaluated here the impact of ERα palmitoylation on E2-induced ERα degradation and S118 phosphorylation. The lack of palmitoylation renders ERα more susceptible to E2-dependent degradation, blocks ERα S118 phosphorylation and prevents E2-induced ERα estrogen-responsive element-containing promoter occupancy. Consequently, ERα transcriptional activity is prevented and the receptor addressed to the nuclear matrix subnuclear compartment. These data uncover a circuitry in which receptor palmitoylation links E2-dependent ERα degradation, S118 phosphorylation, and transcriptional activity in a unique molecular mechanism. We propose that rapid E2-dependent signaling could be considered as a prerequisite for ERα transcriptional activity and suggest an integrated model of ERα intracellular signaling where E2-dependent early extranuclear effects control late receptor-dependent nuclear actions.
Collapse
|
43
|
Anbalagan M, Huderson B, Murphy L, Rowan BG. Post-translational modifications of nuclear receptors and human disease. NUCLEAR RECEPTOR SIGNALING 2012; 10:e001. [PMID: 22438791 PMCID: PMC3309075 DOI: 10.1621/nrs.10001] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 08/19/2011] [Indexed: 12/12/2022]
Abstract
Nuclear receptors (NR) impact a myriad of physiological processes including homeostasis, reproduction, development, and metabolism. NRs are regulated by post-translational modifications (PTM) that markedly impact receptor function. Recent studies have identified NR PTMs that are involved in the onset and progression of human diseases, including cancer. The majority of evidence linking NR PTMs with disease has been demonstrated for phosphorylation, acetylation and sumoylation of androgen receptor (AR), estrogen receptor α (ERα), glucocorticoid receptor (GR) and peroxisome proliferator activated receptor γ (PPARγ). Phosphorylation of AR has been associated with hormone refractory prostate cancer and decreased disease-specific survival. AR acetylation and sumoylation increased growth of prostate cancer tumor models. AR phosphorylation reduced the toxicity of the expanded polyglutamine AR in Kennedy's Disease as a consequence of reduced ligand binding. A comprehensive evaluation of ERα phosphorylation in breast cancer revealed several sites associated with better clinical outcome to tamoxifen therapy, whereas other phosphorylation sites were associated with poorer clinical outcome. ERα acetylation and sumoylation may also have predictive value for breast cancer. GR phosphorylation and acetylation impact GR responsiveness to glucocorticoids that are used as anti-inflammatory drugs. PPARγ phosphorylation can regulate the balance between growth and differentiation in adipose tissue that is linked to obesity and insulin resistance. Sumoylation of PPARγ is linked to repression of inflammatory genes important in patients with inflammatory diseases. NR PTMs provide an additional measure of NR function that can be used as both biomarkers of disease progression, and predictive markers for patient response to NR-directed treatments.
Collapse
Affiliation(s)
- Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | |
Collapse
|
44
|
Regulation of estrogen receptor α N-terminus conformation and function by peptidyl prolyl isomerase Pin1. Mol Cell Biol 2011; 32:445-57. [PMID: 22064478 DOI: 10.1128/mcb.06073-11] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Estrogen receptor alpha (ERα), a key driver of growth in the majority of breast cancers, contains an unstructured transactivation domain (AF1) in its N terminus that is a convergence point for growth factor and hormonal activation. This domain is controlled by phosphorylation, but how phosphorylation impacts AF1 structure and function is unclear. We found that serine 118 (S118) phosphorylation of the ERα AF1 region in response to estrogen (agonist), tamoxifen (antagonist), and growth factors results in recruitment of the peptidyl prolyl cis/trans isomerase Pin1. Phosphorylation of S118 is critical for Pin1 binding, and mutation of S118 to alanine prevents this association. Importantly, Pin1 isomerizes the serine118-proline119 bond from a cis to trans isomer, with a concomitant increase in AF1 transcriptional activity. Pin1 overexpression promotes ligand-independent and tamoxifen-inducible activity of ERα and growth of tamoxifen-resistant breast cancer cells. Pin1 expression correlates with proliferation in ERα-positive rat mammary tumors. These results establish phosphorylation-coupled proline isomerization as a mechanism modulating AF1 functional activity and provide insight into the role of a conformational switch in the functional regulation of the intrinsically disordered transactivation domain of ERα.
Collapse
|
45
|
Yang H, Nguyen TT, An BS, Choi KC, Jeung EB. Synergistic effects of parabens on the induction of calbindin-D9k gene expression act via a progesterone receptor-mediated pathway in GH3 cells. Hum Exp Toxicol 2011; 31:134-44. [DOI: 10.1177/0960327111422402] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although the endocrine-disrupting bioactivity of parabens is weakly estrogenic (parabens are xenoestrogens), their combined synergistic effect is unknown. The aim of this study was to investigate the effects of methyl paraben (MP), ethyl paraben (EP), propyl paraben (PP), isopropyl paraben (IPP), butyl paraben (BP), and isobutyl paraben (IBP), either alone or in combination (MP + EP + PP + BP; PP + IPP; and BP + IBP) on the induction of the estrogenic biomarker gene, calbindin-D9k( CaBP-9k), in rat pituitary lactosomatotrophic GH3 cells. The expression of CaBP-9k mRNA and protein was analyzed using real-time PCR and Western blot analysis, respectively. After 24 h of treatment, a significant increase in CaBP-9k expression was observed. This was dependent upon the length of the paraben alkyl chains (shortest in MP and longest in IBP). Interestingly, the synergistic effects of these paraben combinations were observed at a dose (10−5 M) of these parabens, which induced the highest expression of CaBP-9k mRNA and protein. To investigate the involvement of estrogen receptors (ERs) and progesterone receptors (PRs), through which parabens exert their effects, the expression levels of ERα and PR-B were also examined. The expression of ERα mRNA and protein fluctuated after paraben treatment in GH3 cells, which was not significant. However, the expression level of ERα gene was induced when cotreated with 17β-estradiol (E2) and ICI 182, 780 (estrogen receptor antagonist). The different combinations of parabens induced the expression of the PR-B gene, which was abolished by cotreatment with ICI 182,780. The expression patterns of CaBP-9k and PR-B genes appeared to be similar in response to paraben treatments. This implied that CaBP-9k expression in GH3 cells may be induced by parabens via a PR-mediated pathway. Taken together, these results suggest that exposure to multiple parabens at low concentrations may increase their synergistic estrogenic activities in GH3 cells through a PR-mediated pathway.
Collapse
Affiliation(s)
- H Yang
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - T-T Nguyen
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - B-S An
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - K-C Choi
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - E-B Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
46
|
Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM, Corbo L. Cracking the estrogen receptor's posttranslational code in breast tumors. Endocr Rev 2011; 32:597-622. [PMID: 21680538 DOI: 10.1210/er.2010-0016] [Citation(s) in RCA: 215] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estrogen signaling pathways, because of their central role in regulating the growth and survival of breast tumor cells, have been identified as suitable and efficient targets for cancer therapies. Agents blocking estrogen activity are already widely used clinically, and many new molecules have entered clinical trials, but intrinsic or acquired resistance to treatment limits their efficacy. The basic molecular studies underlying estrogen signaling have defined the critical role of estrogen receptors (ER) in many aspects of breast tumorigenesis. However, important knowledge gaps remain about the role of posttranslational modifications (PTM) of ER in initiation and progression of breast carcinogenesis. Whereas major attention has been focused on the phosphorylation of ER, many other PTM (such as acetylation, ubiquitination, sumoylation, methylation, and palmitoylation) have been identified as events modifying ER expression and stability, subcellular localization, and sensitivity to hormonal response. This article will provide an overview of the current and emerging knowledge on ER PTM, with a particular focus on their deregulation in breast cancer. We also discuss their clinical relevance and the functional relationship between PTM. A thorough understanding of the complete picture of these modifications in ER carcinogenesis might not only open new avenues for identifying new markers for prognosis or prediction of response to endocrine therapy but also could promote the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Muriel Le Romancer
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment Cheney D, 28 rue Laennec, 69373 Lyon Cedex 08, France.
| | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Giamas G, Filipović A, Jacob J, Messier W, Zhang H, Yang D, Zhang W, Shifa BA, Photiou A, Tralau-Stewart C, Castellano L, Green AR, Coombes RC, Ellis IO, Ali S, Lenz HJ, Stebbing J. Kinome screening for regulators of the estrogen receptor identifies LMTK3 as a new therapeutic target in breast cancer. Nat Med 2011; 17:715-9. [PMID: 21602804 DOI: 10.1038/nm.2351] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 03/10/2011] [Indexed: 12/20/2022]
Abstract
Therapies targeting estrogen receptor α (ERα, encoded by ESR1) have transformed the treatment of breast cancer. However, large numbers of women relapse, highlighting the need for the discovery of new regulatory targets modulating ERα pathways. An siRNA screen identified kinases whose silencing alters the estrogen response including those previously implicated in regulating ERα activity (such as mitogen-activated protein kinase and AKT). Among the most potent regulators was lemur tyrosine kinase-3 (LMTK3), for which a role has not previously been assigned. In contrast to other modulators of ERα activity, LMTK3 seems to have been subject to Darwinian positive selection, a noteworthy result given the unique susceptibility of humans to ERα+ breast cancer. LMTK3 acts by decreasing the activity of protein kinase C (PKC) and the phosphorylation of AKT (Ser473), thereby increasing binding of forkhead box O3 (FOXO3) to the ESR1 promoter. LMTK3 phosphorylated ERα, protecting it from proteasomal degradation in vitro. Silencing of LMTK3 reduced tumor volume in an orthotopic mouse model and abrogated proliferation of ERα+ but not ERα- cells, indicative of its role in ERα activity. In human cancers, LMTK3 abundance and intronic polymorphisms were significantly associated with disease-free and overall survival and predicted response to endocrine therapies. These findings yield insights into the natural history of breast cancer in humans and reveal LMTK3 as a new therapeutic target.
Collapse
Affiliation(s)
- Georgios Giamas
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Santin AP, Furlanetto TW. Role of estrogen in thyroid function and growth regulation. J Thyroid Res 2011; 2011:875125. [PMID: 21687614 PMCID: PMC3113168 DOI: 10.4061/2011/875125] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 02/24/2011] [Indexed: 11/23/2022] Open
Abstract
Thyroid diseases are more prevalent in women, particularly between puberty and menopause. It is wellknown that estrogen (E) has indirect effects on the thyroid economy. Direct effects of this steroid hormone on thyroid cells have been described more recently; so, the aim of the present paper was to review the evidences of these effects on thyroid function and growth regulation, and its mechanisms. The expression and ratios of the two E receptors, α and β, that mediate the genomic effects of E on normal and abnormal thyroid tissue were also reviewed, as well as nongenomic, distinct molecular pathways. Several evidences support the hypothesis that E has a direct role in thyroid follicular cells; understanding its influence on the growth and function of the thyroid in normal and abnormal conditions can potentially provide new targets for the treatment of thyroid diseases.
Collapse
Affiliation(s)
- Ana Paula Santin
- Postgraduation Program in Medicine and Medical Sciences, Federal University of Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil
| | | |
Collapse
|
50
|
Ao A, Morrison BJ, Wang H, López JA, Reynolds BA, Lu J. Response of estrogen receptor-positive breast cancer tumorspheres to antiestrogen treatments. PLoS One 2011; 6:e18810. [PMID: 21533195 PMCID: PMC3077404 DOI: 10.1371/journal.pone.0018810] [Citation(s) in RCA: 30] [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: 10/16/2010] [Accepted: 03/18/2011] [Indexed: 12/20/2022] Open
Abstract
Estrogen signaling plays a critical role in the pathogenesis of breast cancer.
Because the majority of breast carcinomas express the estrogen receptor ERα,
endocrine therapy that impedes estrogen-ER signaling reduces breast cancer
mortality and has become a mainstay of breast cancer treatment. However,
patients remain at continued risk of relapse for many years after endocrine
treatment. It has been proposed that cancer recurrence may be attributed to
cancer stem cells (CSCs)/tumor-initiating cells (TICs). Previous studies in
breast cancer have shown that such cells can be enriched and propagated
in vitro by culturing the cells in suspension as
mammospheres/tumorspheres. Here we established tumorspheres from
ERα-positive human breast cancer cell line MCF7 and investigated their
response to antiestrogens Tamoxifen and Fulvestrant. The tumorsphere cells
express lower levels of ERα and are more tumorigenic in xenograft assays
than the parental cells. Both 4-hydroxytamoxifen (4-OHT) and Fulvestrant
attenuate tumorsphere cell proliferation, but only 4-OHT at high concentrations
interferes with sphere formation. However, treated tumorsphere cells retain the
self-renewal capacity. Upon withdrawal of antiestrogens, the treated cells
resume tumorsphere formation and their tumorigenic potential remains undamaged.
Depletion of ERα shows that ERα is dispensable for tumorsphere formation
and xenograft tumor growth in mice. Surprisingly, ERα-depleted tumorspheres
display heightened sensitivity to 4-OHT and their sphere-forming capacity is
diminished after the drug is removed. These results imply that 4-OHT may inhibit
cellular targets besides ERα that are essential for tumorsphere growth, and
provide a potential strategy to sensitize tumorspheres to endocrine
treatment.
Collapse
Affiliation(s)
- Ada Ao
- Department of Biochemistry and Molecular
Biology, University of Florida College of Medicine, Gainesville, Florida, United
States of America
- * E-mail: (JL); (AA)
| | - Brian J. Morrison
- Queensland Institute of Medical Research,
Royal Brisbane Hospital, Brisbane, Australia
- Griffith University, Nathan,
Australia
| | - Heiman Wang
- Department of Biochemistry and Molecular
Biology, University of Florida College of Medicine, Gainesville, Florida, United
States of America
| | - J. Alejandro López
- Queensland Institute of Medical Research,
Royal Brisbane Hospital, Brisbane, Australia
- Griffith University, Nathan,
Australia
| | - Brent A. Reynolds
- Department of Neurosurgery, McKnight Brain
Institute, University of Florida, Gainesville, Florida, United States of
America
| | - Jianrong Lu
- Department of Biochemistry and Molecular
Biology, University of Florida College of Medicine, Gainesville, Florida, United
States of America
- * E-mail: (JL); (AA)
| |
Collapse
|