1
|
Harrod A, Lai CF, Goldsbrough I, Simmons GM, Oppermans N, Santos DB, Győrffy B, Allsopp RC, Toghill BJ, Balachandran K, Lawson M, Morrow CJ, Surakala M, Carnevalli LS, Zhang P, Guttery DS, Shaw JA, Coombes RC, Buluwela L, Ali S. Genome engineering for estrogen receptor mutations reveals differential responses to anti-estrogens and new prognostic gene signatures for breast cancer. Oncogene 2022; 41:4905-4915. [PMID: 36198774 PMCID: PMC7613769 DOI: 10.1038/s41388-022-02483-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022]
Abstract
Mutations in the estrogen receptor (ESR1) gene are common in ER-positive breast cancer patients who progress on endocrine therapies. Most mutations localise to just three residues at, or near, the C-terminal helix 12 of the hormone binding domain, at leucine-536, tyrosine-537 and aspartate-538. To investigate these mutations, we have used CRISPR-Cas9 mediated genome engineering to generate a comprehensive set of isogenic mutant breast cancer cell lines. Our results confirm that L536R, Y537C, Y537N, Y537S and D538G mutations confer estrogen-independent growth in breast cancer cells. Growth assays show mutation-specific reductions in sensitivities to drugs representing three classes of clinical anti-estrogens. These differential mutation- and drug-selectivity profiles have implications for treatment choices following clinical emergence of ER mutations. Our results further suggest that mutant expression levels may be determinants of the degree of resistance to some anti-estrogens. Differential gene expression analysis demonstrates up-regulation of estrogen-responsive genes, as expected, but also reveals that enrichment for interferon-regulated gene expression is a common feature of all mutations. Finally, a new gene signature developed from the gene expression profiles in ER mutant cells predicts clinical response in breast cancer patients with ER mutations.
Collapse
Affiliation(s)
- Alison Harrod
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK
- Institute of Cancer Research, Fulham Road, London, SW3 6JB, UK
| | - Chun-Fui Lai
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK
| | | | - Georgia M Simmons
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK
| | - Natasha Oppermans
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK
| | - Daniela B Santos
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK
| | - Balazs Győrffy
- Semmelweis University Department of Bioinformatics, H-1094 Budapest, Hungary and TTK Cancer Biomarker Research Group, H-1117, Budapest, Hungary
| | - Rebecca C Allsopp
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK
| | - Bradley J Toghill
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK
| | - Kirsty Balachandran
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK
| | - Mandy Lawson
- Early Oncology R&D, AstraZeneca, Biomedical Campus, 1 Francis Crick Ave, Cambridge, CB2 0AA, UK
| | - Christopher J Morrow
- Early Oncology R&D, AstraZeneca, Biomedical Campus, 1 Francis Crick Ave, Cambridge, CB2 0AA, UK
| | - Manasa Surakala
- Early Oncology R&D, AstraZeneca, Biomedical Campus, 1 Francis Crick Ave, Cambridge, CB2 0AA, UK
| | - Larissa S Carnevalli
- Early Oncology R&D, AstraZeneca, Biomedical Campus, 1 Francis Crick Ave, Cambridge, CB2 0AA, UK
| | - Pei Zhang
- Early Oncology R&D, AstraZeneca, Biomedical Campus, 1 Francis Crick Ave, Cambridge, CB2 0AA, UK
| | - David S Guttery
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK
| | - Jacqueline A Shaw
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK
| | - R Charles Coombes
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK
| | - Lakjaya Buluwela
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK.
| | - Simak Ali
- Department of Surgery & Cancer, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
2
|
Arao Y, Korach KS. The physiological role of estrogen receptor functional domains. Essays Biochem 2021; 65:867-875. [PMID: 34028522 PMCID: PMC8611119 DOI: 10.1042/ebc20200167] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/21/2021] [Accepted: 05/07/2021] [Indexed: 01/27/2023]
Abstract
Estrogen receptor (ER) is a member of the nuclear receptor superfamily whose members share conserved domain structures, including a DNA-binding domain (DBD) and ligand-binding domain (LBD). Estrogenic chemicals work as ligands for activation or repression of ER-mediated transcriptional activity derived from two transactivation domains: AF-1 and AF-2. AF-2 is localized in the LBD, and helix 12 of the LBD is essential for controlling AF-2 functionality. The positioning of helix 12 defines the ER alpha (ERα) ligand properties as agonists or antagonists. In contrast, it is still less well defined as to the ligand-dependent regulation of N-terminal AF-1 activity. It has been thought that the action of selective estrogen receptor modulators (SERMs) is mediated by the regulation of a tissue specific AF-1 activity rather than AF-2 activity. However, it is still unclear how SERMs regulate AF-1 activity in a tissue-selective manner. This review presents some recent observations toward information of ERα mediated SERM actions related to the ERα domain functionality, focusing on the following topics. (1) The F-domain, which is connected to helix 12, controls 4-hydroxytamoxifen (4OHT) mediated AF-1 activation associated with the receptor dimerization activity. (2) The zinc-finger property of the DBD for genomic sequence recognition. (3) The novel estrogen responsive genomic DNA element, which contains multiple long-spaced direct-repeats without a palindromic ERE sequence, is differentially recognized by 4OHT and E2 ligand bound ERα transactivation complexes.
Collapse
Affiliation(s)
- Yukitomo Arao
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH
| | - Kenneth S Korach
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH
| |
Collapse
|
3
|
Katzenellenbogen JA, Mayne CG, Katzenellenbogen BS, Greene GL, Chandarlapaty S. Structural underpinnings of oestrogen receptor mutations in endocrine therapy resistance. Nat Rev Cancer 2018; 18:377-388. [PMID: 29662238 PMCID: PMC6252060 DOI: 10.1038/s41568-018-0001-z] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oestrogen receptor-α (ERα), a key driver of breast cancer, normally requires oestrogen for activation. Mutations that constitutively activate ERα without the need for hormone binding are frequently found in endocrine-therapy-resistant breast cancer metastases and are associated with poor patient outcomes. The location of these mutations in the ER ligand-binding domain and their impact on receptor conformation suggest that they subvert distinct mechanisms that normally maintain the low basal state of wild-type ERα in the absence of hormone. Such mutations provide opportunities to probe fundamental issues underlying ligand-mediated control of ERα activity. Instructive contrasts between these ERα mutations and those that arise in the androgen receptor (AR) during anti-androgen treatment of prostate cancer highlight differences in how activation functions in ERs and AR control receptor activity, how hormonal pressures (deprivation versus antagonism) drive the selection of phenotypically different mutants, how altered protein conformations can reduce antagonist potency and how altered ligand-receptor contacts can invert the response that a receptor has to an agonist ligand versus an antagonist ligand. A deeper understanding of how ligand regulation of receptor conformation is linked to receptor function offers a conceptual framework for developing new anti-oestrogens that might be more effective in preventing and treating breast cancer.
Collapse
Affiliation(s)
| | - Christopher G Mayne
- Beckman Institute for Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Geoffrey L Greene
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
4
|
Harrod A, Fulton J, Nguyen VTM, Periyasamy M, Ramos-Garcia L, Lai CF, Metodieva G, de Giorgio A, Williams RL, Santos DB, Gomez PJ, Lin ML, Metodiev MV, Stebbing J, Castellano L, Magnani L, Coombes RC, Buluwela L, Ali S. Genomic modelling of the ESR1 Y537S mutation for evaluating function and new therapeutic approaches for metastatic breast cancer. Oncogene 2017; 36:2286-2296. [PMID: 27748765 PMCID: PMC5245767 DOI: 10.1038/onc.2016.382] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/23/2016] [Accepted: 08/29/2016] [Indexed: 02/07/2023]
Abstract
Drugs that inhibit estrogen receptor-α (ER) activity have been highly successful in treating and reducing breast cancer progression in ER-positive disease. However, resistance to these therapies presents a major clinical problem. Recent genetic studies have shown that mutations in the ER gene are found in >20% of tumours that progress on endocrine therapies. Remarkably, the great majority of these mutations localize to just a few amino acids within or near the critical helix 12 region of the ER hormone binding domain, where they are likely to be single allele mutations. Understanding how these mutations impact on ER function is a prerequisite for identifying methods to treat breast cancer patients featuring such mutations. Towards this end, we used CRISPR-Cas9 genome editing to make a single allele knock-in of the most commonly mutated amino acid residue, tyrosine 537, in the estrogen-responsive MCF7 breast cancer cell line. Genomic analyses using RNA-seq and ER ChIP-seq demonstrated that the Y537S mutation promotes constitutive ER activity globally, resulting in estrogen-independent growth. MCF7-Y537S cells were resistant to the anti-estrogen tamoxifen and fulvestrant. Further, we show that the basal transcription factor TFIIH is constitutively recruited by ER-Y537S, resulting in ligand-independent phosphorylation of Serine 118 (Ser118) by the TFIIH kinase, cyclin-dependent kinase (CDK)7. The CDK7 inhibitor, THZ1 prevented Ser118 phosphorylation and inhibited growth of MCF7-Y537S cells. These studies confirm the functional importance of ER mutations in endocrine resistance, demonstrate the utility of knock-in mutational models for investigating alternative therapeutic approaches and highlight CDK7 inhibition as a potential therapy for endocrine-resistant breast cancer mediated by ER mutations.
Collapse
Affiliation(s)
- A Harrod
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - J Fulton
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - V T M Nguyen
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - M Periyasamy
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - L Ramos-Garcia
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - C-F Lai
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - G Metodieva
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, UK
| | - A de Giorgio
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - R L Williams
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - D B Santos
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - P J Gomez
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - M-L Lin
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - M V Metodiev
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester, UK
| | - J Stebbing
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - L Castellano
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - L Magnani
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - R C Coombes
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - L Buluwela
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - S Ali
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
5
|
Varešlija D, McBryan J, Fagan A, Redmond AM, Hao Y, Sims AH, Turnbull A, Dixon JM, Ó Gaora P, Hudson L, Purcell S, Hill ADK, Young LS. Adaptation to AI Therapy in Breast Cancer Can Induce Dynamic Alterations in ER Activity Resulting in Estrogen-Independent Metastatic Tumors. Clin Cancer Res 2016; 22:2765-77. [PMID: 26763249 DOI: 10.1158/1078-0432.ccr-15-1583] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/20/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Acquired resistance to aromatase inhibitor (AI) therapy is a major clinical problem in the treatment of breast cancer. The detailed mechanisms of how tumor cells develop this resistance remain unclear. Here, the adapted function of estrogen receptor (ER) to an estrogen-depleted environment following AI treatment is reported. EXPERIMENTAL DESIGN Global ER chromatin immuno-precipitation (ChIP)-seq analysis of AI-resistant cells identified steroid-independent ER target genes. Matched patient tumor samples, collected before and after AI treatment, were used to assess ER activity. RESULTS Maintained ER activity was observed in patient tumors following neoadjuvant AI therapy. Genome-wide ER-DNA-binding analysis in AI-resistant cell lines identified a subset of classic ligand-dependent ER target genes that develop steroid independence. The Kaplan-Meier analysis revealed a significant association between tumors, which fail to decrease this steroid-independent ER target gene set in response to neoadjuvant AI therapy, and poor disease-free survival and overall survival (n = 72 matched patient tumor samples, P = 0.00339 and 0.00155, respectively). The adaptive ER response to AI treatment was highlighted by the ER/AIB1 target gene, early growth response 3 (EGR3). Elevated levels of EGR3 were detected in endocrine-resistant local disease recurrent patient tumors in comparison with matched primary tissue. However, evidence from distant metastatic tumors demonstrates that the ER signaling network may undergo further adaptations with disease progression as estrogen-independent ER target gene expression is routinely lost in established metastatic tumors. CONCLUSIONS Overall, these data provide evidence of a dynamic ER response to endocrine treatment that may provide vital clues for overcoming the clinical issue of therapy resistance. Clin Cancer Res; 22(11); 2765-77. ©2016 AACR.
Collapse
Affiliation(s)
- Damir Varešlija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jean McBryan
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ailís Fagan
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Aisling M Redmond
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Andrew H Sims
- University of Edinburgh Cancer Research Centre, Carrington Crescent, Edinburgh, EH4 2XU, United Kingdom
| | - Arran Turnbull
- University of Edinburgh Cancer Research Centre, Carrington Crescent, Edinburgh, EH4 2XU, United Kingdom
| | - J M Dixon
- University of Edinburgh Cancer Research Centre, Carrington Crescent, Edinburgh, EH4 2XU, United Kingdom
| | - Peadar Ó Gaora
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Lance Hudson
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Siobhan Purcell
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Arnold D K Hill
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Leonie S Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| |
Collapse
|
6
|
Hoffmann JM, Partridge L. Nuclear hormone receptors: Roles of xenobiotic detoxification and sterol homeostasis in healthy aging. Crit Rev Biochem Mol Biol 2015; 50:380-92. [PMID: 26383043 DOI: 10.3109/10409238.2015.1067186] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Health during aging can be improved by genetic, dietary and pharmacological interventions. Many of these increase resistance to various stressors, including xenobiotics. Up-regulation of xenobiotic detoxification genes is a transcriptomic signature shared by long-lived nematodes, flies and mice, suggesting that protection of cells from toxicity of xenobiotics may contribute to longevity. Expression of genes involved in xenobiotic detoxification is controlled by evolutionarily conserved transcriptional regulators. Three closely related subgroups of nuclear hormone receptors (NHRs) have a major role, and these include DAF-12 and NHR-8 in C. elegans, DHR96 in Drosophila and FXR, LXRs, PXR, CAR and VDR in mammals. In the invertebrates, these NHRs have been experimentally demonstrated to play a role in extension of lifespan by genetic and environmental interventions. NHRs represent critical hubs in that they regulate detoxification enzymes with broad substrate specificities, metabolizing both endo- and xeno-biotics. They also modulate homeostasis of steroid hormones and other endogenous cholesterol derivatives and lipid metabolism, and these roles, as well as xenobiotic detoxification, may contribute to the effects of NHRs on lifespan and health during aging, an issue that is being increasingly addressed in C. elegans and Drosophila. Disentangling the contribution of these processes to longevity will require more precise understanding of the molecular mechanisms by which each is effected, including identification of ligands and co-regulators of NHRs, patterns of tissue-specificity and mechanisms of interaction between tissues. The roles of vertebrate NHRs in determination of health during aging and lifespan have yet to be investigated.
Collapse
Affiliation(s)
| | - Linda Partridge
- a Max Planck Institute for Biology of Ageing , Cologne , Germany and.,b Institute of Healthy Ageing, and GEE (Genetics, Evolution and Environment), University College , London , UK
| |
Collapse
|
7
|
Thomas C, Gustafsson JÅ. Estrogen receptor mutations and functional consequences for breast cancer. Trends Endocrinol Metab 2015; 26:467-76. [PMID: 26183887 DOI: 10.1016/j.tem.2015.06.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 02/07/2023]
Abstract
A significant number of estrogen receptor α (ERα)-positive breast tumors develop resistance to endocrine therapy and recur with metastatic disease. Several mechanisms of endocrine resistance have been proposed, including genetic alterations that lead to ERs with altered protein sequence. By altering the conformation of the protein and increasing the interaction with coactivators, point mutations in ESR1, the gene encoding ERα, promote an active form of the receptor in the absence of hormone that assists tumor cells to evade hormonal treatments. Recent studies have confirmed that ESR1 point mutations frequently occur in metastatic breast tumors that are refractory to endocrine therapy, and suggest the development of novel strategies that may be more effective in controlling ER signaling and benefit patients with recurrent and metastatic disease.
Collapse
Affiliation(s)
- Christoforos Thomas
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Boulevard, Houston, TX 77204, USA.
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Boulevard, Houston, TX 77204, USA.
| |
Collapse
|
8
|
Tharun IM, Nieto L, Haase C, Scheepstra M, Balk M, Möcklinghoff S, Adriaens W, Dames SA, Brunsveld L. Subtype-specific modulation of estrogen receptor-coactivator interaction by phosphorylation. ACS Chem Biol 2015; 10:475-84. [PMID: 25386784 DOI: 10.1021/cb5007097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The estrogen receptor (ER) is the number one target for the treatment of endocrine responsive breast cancer and remains a highly attractive target for new drug development. Despite considerable efforts to understand the role of ER post-translational modifications (PTMs), the complexity of these modifications and their impact, at the molecular level, are poorly understood. Using a chemical biology approach, fundamentally rooted in an efficient protein semisynthesis of tyrosine phosphorylated ER constructs, the complex role of the ER tyrosine phosphorylation is addressed here for the first time on a molecular level. The semisynthetic approach allows for the site-specific introduction of PTMs as well as biophysical probes. A combination of biophysical techniques, including NMR, with molecular dynamics studies reveals the role of the phosphorylation of the clinically relevant tyrosine 537 (Y537) in ERα and the analogous tyrosine (Y488) in ERβ. Phosphorylation has important effects on the dynamics of the ER Helix 12, which is centrally involved in receptor activity regulation, and on its interplay with ligand and cofactor binding, but with differential regulatory effects of the analogous PTMs on the two ER subtypes. Combined, the results bring forward a novel molecular model of a phosphorylation-induced subtype specific ER modulatory mechanism, alternative to the widely accepted ligand-induced activation mechanism.
Collapse
Affiliation(s)
- Inga M. Tharun
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| | - Lidia Nieto
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| | - Christian Haase
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| | - Marcel Scheepstra
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| | - Mark Balk
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| | - Sabine Möcklinghoff
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| | - Wencke Adriaens
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| | - Sonja A. Dames
- Chair
of Biomolecular NMR Spectroscopy, Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
- Institute
of Structural Biology, Helmholtz Zentrum München, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Luc Brunsveld
- Laboratory
of Chemical Biology and Institute of Complex Molecular Systems, Department
of Biomedical Engineering, Eindhoven University of Technology, Den Dolech
2, 5612AZ Eindhoven, The Netherlands
| |
Collapse
|
9
|
Smith KP, Gifford KM, Waitzman JS, Rice SE. Survey of phosphorylation near drug binding sites in the Protein Data Bank (PDB) and their effects. Proteins 2015; 83:25-36. [PMID: 24833420 PMCID: PMC4233198 DOI: 10.1002/prot.24605] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/28/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
While it is currently estimated that 40 to 50% of eukaryotic proteins are phosphorylated, little is known about the frequency and local effects of phosphorylation near pharmaceutical inhibitor binding sites. In this study, we investigated how frequently phosphorylation may affect the binding of drug inhibitors to target proteins. We examined the 453 non-redundant structures of soluble mammalian drug target proteins bound to inhibitors currently available in the Protein Data Bank (PDB). We cross-referenced these structures with phosphorylation data available from the PhosphoSitePlus database. Three hundred twenty-two of 453 (71%) of drug targets have evidence of phosphorylation that has been validated by multiple methods or labs. For 132 of 453 (29%) of those, the phosphorylation site is within 12 Å of the small molecule-binding site, where it would likely alter small molecule binding affinity. We propose a framework for distinguishing between drug-phosphorylation site interactions that are likely to alter the efficacy of drugs versus those that are not. In addition we highlight examples of well-established drug targets, such as estrogen receptor alpha, for which phosphorylation may affect drug affinity and clinical efficacy. Our data suggest that phosphorylation may affect drug binding and efficacy for a significant fraction of drug target proteins.
Collapse
Affiliation(s)
- Kyle P Smith
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611
| | | | | | | |
Collapse
|
10
|
Bridgham JT, Keay J, Ortlund EA, Thornton JW. Vestigialization of an allosteric switch: genetic and structural mechanisms for the evolution of constitutive activity in a steroid hormone receptor. PLoS Genet 2014; 10:e1004058. [PMID: 24415950 PMCID: PMC3886901 DOI: 10.1371/journal.pgen.1004058] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 11/08/2013] [Indexed: 11/30/2022] Open
Abstract
An important goal in molecular evolution is to understand the genetic and physical mechanisms by which protein functions evolve and, in turn, to characterize how a protein's physical architecture influences its evolution. Here we dissect the mechanisms for an evolutionary shift in function in the mollusk ortholog of the steroid hormone receptors (SRs), a family of biologically essential transcription factors. In vertebrates, the activity of SRs allosterically depends on binding a hormonal ligand; in mollusks, however, the SR ortholog (called ER, because of high sequence similarity to vertebrate estrogen receptors) activates transcription in the absence of ligand and does not respond to steroid hormones. To understand how this shift in regulation evolved, we combined evolutionary, structural, and functional analyses. We first determined the X-ray crystal structure of the ER of the Pacific oyster Crassostrea gigas (CgER), and found that its ligand pocket is filled with bulky residues that prevent ligand occupancy. To understand the genetic basis for the evolution of mollusk ERs' unique functions, we resurrected an ancient SR progenitor and characterized the effect of historical amino acid replacements on its functions. We found that reintroducing just two ancient replacements from the lineage leading to mollusk ERs recapitulates the evolution of full constitutive activity and the loss of ligand activation. These substitutions stabilize interactions among key helices, causing the allosteric switch to become “stuck” in the active conformation and making activation independent of ligand binding. Subsequent changes filled the ligand pocket without further affecting activity; by degrading the allosteric switch, these substitutions vestigialized elements of the protein's architecture required for ligand regulation and made reversal to the ancestral function more complex. These findings show how the physical architecture of allostery enabled a few large-effect mutations to trigger a profound evolutionary change in the protein's function and shaped the genetics of evolutionary reversibility. An important goal in evolutionary genetics is to understand how genetic mutations cause the evolution of new protein functions and how a protein's structure shapes its evolution. Here we address these questions by studying a dramatic lineage-specific shift in function in steroid hormone receptors (SRs), a physiologically important family of transcription factors. In vertebrates, SRs bind hormones and then undergo a structural change that allows them to activate gene expression. In mollusks, SRs do not bind hormone and are always active. We identified the genetic and structural mechanisms for the evolution of constitutive activity in the mollusk SRs by using X-ray crystallography, ancestral sequence reconstruction, and experimental studies of the effects of ancient mutations on protein structure and function. We found that constitutive activity evolved due to just two historical substitutions that subtly stabilized elements of the active conformation, and subsequent mutations filled the hormone-binding cavity. The structural characteristics required for a hormone-sensitive activator were thus vestigialized, much the same way that a whale's hindlimbs became vestiges of their ancestral form after they became dispensable. Our findings show how the architecture of a protein can shape its evolution, allowing radically different functions to evolve by a few large-effect mutations.
Collapse
Affiliation(s)
- Jamie T. Bridgham
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon, United States of America
| | - June Keay
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon, United States of America
| | - Eric A. Ortlund
- Biochemistry Department, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Joseph W. Thornton
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon, United States of America
- Departments of Human Genetics and Ecology & Evolution, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
11
|
Jeselsohn R, Yelensky R, Buchwalter G, Frampton G, Meric-Bernstam F, Gonzalez-Angulo AM, Ferrer-Lozano J, Perez-Fidalgo JA, Cristofanilli M, Gómez H, Arteaga CL, Giltnane J, Balko JM, Cronin MT, Jarosz M, Sun J, Hawryluk M, Lipson D, Otto G, Ross JS, Dvir A, Soussan-Gutman L, Wolf I, Rubinek T, Gilmore L, Schnitt S, Come SE, Pusztai L, Stephens P, Brown M, Miller VA. Emergence of constitutively active estrogen receptor-α mutations in pretreated advanced estrogen receptor-positive breast cancer. Clin Cancer Res 2014; 20:1757-1767. [PMID: 24398047 DOI: 10.1158/1078-0432.ccr-13-2332] [Citation(s) in RCA: 503] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE We undertook this study to determine the prevalence of estrogen receptor (ER) α (ESR1) mutations throughout the natural history of hormone-dependent breast cancer and to delineate the functional roles of the most commonly detected alterations. EXPERIMENTAL DESIGN We studied a total of 249 tumor specimens from 208 patients. The specimens include 134 ER-positive (ER(+)/HER2(-)) and, as controls, 115 ER-negative (ER(-)) tumors. The ER(+) samples consist of 58 primary breast cancers and 76 metastatic samples. All tumors were sequenced to high unique coverage using next-generation sequencing targeting the coding sequence of the estrogen receptor and an additional 182 cancer-related genes. RESULTS Recurring somatic mutations in codons 537 and 538 within the ligand-binding domain of ER were detected in ER(+) metastatic disease. Overall, the frequency of these mutations was 12% [9/76; 95% confidence interval (CI), 6%-21%] in metastatic tumors and in a subgroup of patients who received an average of 7 lines of treatment the frequency was 20% (5/25; 95% CI, 7%-41%). These mutations were not detected in primary or treatment-naïve ER(+) cancer or in any stage of ER(-) disease. Functional studies in cell line models demonstrate that these mutations render estrogen receptor constitutive activity and confer partial resistance to currently available endocrine treatments. CONCLUSIONS In this study, we show evidence for the temporal selection of functional ESR1 mutations as potential drivers of endocrine resistance during the progression of ER(+) breast cancer.
Collapse
Affiliation(s)
- Rinath Jeselsohn
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, 450 Brookline Ave. Boston, MA 02215.,Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215
| | - Roman Yelensky
- Foundation Medicine, One Kendall Sq. Cambridge, MA 02139
| | - Gilles Buchwalter
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, 450 Brookline Ave. Boston, MA 02215.,Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215
| | | | - Funda Meric-Bernstam
- Departments of Investigational Cancer Therapeutics, Surgical Oncology, The University of MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Ana Maria Gonzalez-Angulo
- Departments of Systems Biology, and Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Jaime Ferrer-Lozano
- Fundacion de Investigacion INCLIVA - Institute for Health Reseearch, Valencia, Spain
| | - Jose A Perez-Fidalgo
- Departments of Hematology-Oncology, Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - Massimo Cristofanilli
- Jefferson Breast Care Center, Kimmel Cancer Center, Thomas Jefferson University, 925 Chestnut St. Philadelphia, PA 19107
| | - Henry Gómez
- Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Perú
| | - Carlos L Arteaga
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN 37232
| | - Jennifer Giltnane
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN 37232
| | - Justin M Balko
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN 37232
| | | | - Mirna Jarosz
- Foundation Medicine, One Kendall Sq. Cambridge, MA 02139
| | - James Sun
- Foundation Medicine, One Kendall Sq. Cambridge, MA 02139
| | | | - Doron Lipson
- Foundation Medicine, One Kendall Sq. Cambridge, MA 02139
| | - Geoff Otto
- Foundation Medicine, One Kendall Sq. Cambridge, MA 02139
| | - Jeffrey S Ross
- Foundation Medicine, One Kendall Sq. Cambridge, MA 02139
| | - Addie Dvir
- Teva Pharmaceuticals, 5 Basel St. Petach Tikva, Israel 49131
| | | | - Ido Wolf
- Oncology Division, Tel Aviv Sourasky Medical Center , 6 Weizmann St. Tel Aviv 64239, Israel
| | - Tamar Rubinek
- Oncology Division, Tel Aviv Sourasky Medical Center , 6 Weizmann St. Tel Aviv 64239, Israel
| | - Lauren Gilmore
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave. Boston MA 02215
| | - Stuart Schnitt
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave. Boston MA 02215
| | - Steven E Come
- Breast Medical Oncology Program, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave. Boston MA 02215
| | - Lajos Pusztai
- Section of Breast Medical Oncology, Yale School of Medicine, New Haven, South Frontage Rd and Park St. CN, 06510
| | | | - Myles Brown
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, 450 Brookline Ave. Boston, MA 02215.,Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215
| | | |
Collapse
|
12
|
Slavin S, Yeh CR, Da J, Yu S, Miyamoto H, Messing EM, Guancial E, Yeh S. Estrogen receptor α in cancer-associated fibroblasts suppresses prostate cancer invasion via modulation of thrombospondin 2 and matrix metalloproteinase 3. Carcinogenesis 2013; 35:1301-9. [PMID: 24374826 DOI: 10.1093/carcin/bgt488] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The prostate cancer (PCa) microenvironment contains active stromal cells known as cancer-associated fibroblasts (CAF) that may play important roles in influencing tumor progression. Here we studied the role of CAF estrogen receptor alpha (ERα) and found that it could protect against PCa invasion. Immunohistochemistry on prostatectomy specimens showed that PCa patients with ERα-positive stroma had a significantly lower risk for biochemical recurrence. In vitro invasion assays further confirmed that the stromal ERα was able to reduce PCa cell invasion. Dissection of the molecular mechanism revealed that the CAF ERα could function through a CAF-epithelial interaction via selectively upregulating thrombospondin 2 (Thbs2) and downregulating matrix metalloproteinase 3 (MMP3) at the protein and messenger RNA levels. Chromatin immunoprecipitation assays further showed that ERα could bind to an estrogen response element on the promoter of Thbs2. Importantly, knockdown of Thbs2 led to increased MMP3 expression and interruption of the ERα mediated invasion suppression, providing further evidence of an ERα-Thbs2-MMP3 axis in CAF. In vivo studies using athymic nude mice injected with CWR22Rv1 (22Rv1) PCa epithelial cells and CAF cells ± ERα also confirmed that mice coimplanted with PCa cells and CAF ERα+ cells had less tumor foci in the pelvic lymph nodes, less metastases, and tumors showed less angiogenesis, MMP3, and MMP9 (an MMP3 downstream target) positive staining. Together, these data suggest that CAF ERα could play protective roles in suppressing PCa metastasis. Our results may lead to developing new and alternative therapeutic approaches to battle PCa via controlling ERα signaling in CAF.
Collapse
Affiliation(s)
- Spencer Slavin
- Departments of Urology and Pathology, University of Rochester Medical Center Rochester, 601 Elmwood Avenue, NY 14642, USA
| | - Chiuan-Ren Yeh
- Departments of Urology and Pathology, University of Rochester Medical Center Rochester, 601 Elmwood Avenue, NY 14642, USA
| | - Jun Da
- Departments of Urology and Pathology, University of Rochester Medical Center Rochester, 601 Elmwood Avenue, NY 14642, USA, Department of Urology, Shanghai Jaotong University, Shanghai, China and
| | - Shengqiang Yu
- Departments of Urology and Pathology, University of Rochester Medical Center Rochester, 601 Elmwood Avenue, NY 14642, USA
| | - Hiroshi Miyamoto
- Departments of Urology and Pathology, University of Rochester Medical Center Rochester, 601 Elmwood Avenue, NY 14642, USA
| | - Edward M Messing
- Departments of Urology and Pathology, University of Rochester Medical Center Rochester, 601 Elmwood Avenue, NY 14642, USA
| | - Elizabeth Guancial
- Departments of Hematology and Oncology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Shuyuan Yeh
- Departments of Urology and Pathology, University of Rochester Medical Center Rochester, 601 Elmwood Avenue, NY 14642, USA,
| |
Collapse
|
13
|
Ferriere F, Habauzit D, Pakdel F, Saligaut C, Flouriot G. Unliganded estrogen receptor alpha promotes PC12 survival during serum starvation. PLoS One 2013; 8:e69081. [PMID: 23825704 PMCID: PMC3692477 DOI: 10.1371/journal.pone.0069081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 06/10/2013] [Indexed: 12/24/2022] Open
Abstract
Many studies have reported proliferative, differentiating or protective effects of estradiol, notably through estrogen receptor alpha (ERα). On the contrary, the ligand-independent action of ERα is currently poorly documented notably in cell protection. The stable transfection of wild type, substituted or truncated form of ERα in PC12 cells (ERα negative cell line) lead the specific study of its ligand-independent action. Hence, we demonstrate here that, in the absence of E2, the expression of ERα prevents cells from apoptosis induced by serum deprivation. This protection is not due to an ERE-mediated transcription and does not require either AF-1 or AF-2 transactivation functions. It is afforded to the Y537 residue of ERα and activation of c-Src/Stat3 signaling pathway.
Collapse
Affiliation(s)
- François Ferriere
- Transcription, Environment and Cancer Group, Institut de Recherche sur la Santé, Environnement et Travail (IRSET), Institut National de la Santé et de la Recherche Médicale (INSERM) U1085, Université de Rennes 1, Rennes, France.
| | | | | | | | | |
Collapse
|
14
|
Castoria G, Giovannelli P, Lombardi M, De Rosa C, Giraldi T, de Falco A, Barone MV, Abbondanza C, Migliaccio A, Auricchio F. Tyrosine phosphorylation of estradiol receptor by Src regulates its hormone-dependent nuclear export and cell cycle progression in breast cancer cells. Oncogene 2012; 31:4868-77. [PMID: 22266855 DOI: 10.1038/onc.2011.642] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 12/13/2011] [Accepted: 12/16/2011] [Indexed: 12/23/2022]
Abstract
We report that in breast cancer cells, tyrosine phosphorylation of the estradiol receptor alpha (ERalpha) by Src regulates cytoplasmic localization of the receptor and DNA synthesis. Inhibition of Src or use of a peptide mimicking the ERalpha p-Tyr537 sequence abolishes ERalpha tyrosine phosphorylation and traps the receptor in nuclei of estradiol-treated MCF-7 cells. An ERalpha mutant carrying a mutation of Tyr537 to phenylalanine (ER537F) persistently localizes in nuclei of various cell types. In contrast with ERalpha wt, ER537F does not associate with Ran and its interaction with Crm1 is insensitive to estradiol. Thus, independently of estradiol, ER537F is retained in nuclei, where it entangles FKHR-driving cell cycle arrest. Chromatin immunoprecipitation analysis reveals that overexpression of ER537F in breast cancer cells enhances FKHR interaction with cyclin D1 promoter. This mutant also counteracts cell transformation by the activated forms of Src or PI3-K. In conclusion, in addition to regulating receptor localization, ERalpha phosphorylation by Src is required for hormone responsiveness of DNA synthesis in breast cancer cells.
Collapse
Affiliation(s)
- G Castoria
- 1] Department of General Pathology, II University of Naples, Naples, Italy [2] These authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Development of a novel molecular sensor for imaging estrogen receptor-coactivator protein-protein interactions. PLoS One 2012; 7:e44160. [PMID: 22952913 PMCID: PMC3429467 DOI: 10.1371/journal.pone.0044160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/30/2012] [Indexed: 11/21/2022] Open
Abstract
Anti-estrogens, in particular tissue selective anti-estrogens, have been the bedrock of adjuvant therapy for patients with estrogen receptor alpha (ERα) positive breast cancer. Though current therapies have greatly enhanced patient prognosis, there continues to be an impetus for the development of improved anti-estrogens. ERα is a nuclear receptor transcription factor which activates gene expression through the recruitment of transcriptional coactivator proteins. The SRC family of coactivators, which includes AIB1, has been shown to be of particular importance for ERα mediated transcription. ERα-AIB1 interactions are indicative of gene expression and are inhibited by anti-estrogen treatment. We have exploited the interaction between ERα and AIB1 as a novel method for imaging ERα activity using a split luciferase molecular sensor. By producing a range of ERα ligand binding domain (ER-LBD) and AIB1 nuclear receptor interacting domain (AIB-RID) N- and C-terminal firefly luciferase fragment fusion proteins, constructs which exhibited more than a 10-fold increase in luciferase activity with E2 stimulation were identified. The specificity of the E2-stimulated luciferase activity to ERα-AIB1 interaction was validated through Y537S and L539/540A ER-LBD fusion protein mutants. The primed nature of the split luciferase assay allowed changes in ERα activity, with respect to the protein-protein interactions preceding transcription, to be assessed soon after drug treatment. The novel assay split luciferase detailed in this report enabled modulation of ERα activity to be sensitively imaged in vitro and in living subjects and potentially holds much promise for imaging the efficacy of novel ERα specific therapies.
Collapse
|
16
|
Paisley JC, Huddleston GG, Carruth LL, Petrulis A, Grober MS, Clancy AN. Sexual responses of the male rat medial preoptic area and medial amygdala to estrogen I: site specific suppression of estrogen receptor alpha. Horm Behav 2012; 62:50-7. [PMID: 22565217 DOI: 10.1016/j.yhbeh.2012.04.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/31/2012] [Accepted: 04/28/2012] [Indexed: 11/17/2022]
Abstract
Male rat copulation is mediated by estrogen-sensitive neurons in the medial preoptic area (MPO) and medial amygdala (MEA); however, the mechanisms through which estradiol (E(2)) acts are not fully understood. We hypothesized that E(2) acts through estrogen receptor α (ERα) in the MPO and MEA to promote male mating behavior. Antisense oligodeoxynucleotides (AS-ODN) complementary to ERα mRNA were bilaterally infused via minipumps into either brain area to block the synthesis of ERα, which we predicted would reduce mating. Western blot analysis and immunocytochemistry revealed a knockdown of ERα expression in each brain region; however, compared to saline controls, males receiving AS-ODN to the MPO showed significant reductions in all components of mating, whereas males receiving AS-ODN to the MEA continued to mate normally. These results suggest that E(2) acts differently in these brain regions to promote the expression of male rat sexual behavior and that ERα in the MPO, but not in the MEA, promotes mating.
Collapse
Affiliation(s)
- Jacquelyn C Paisley
- Department of Biology, Georgia State University, Atlanta, GA 30302‐4010, USA
| | | | | | | | | | | |
Collapse
|
17
|
Bernardo TJ, Dubrovsky EB. Molecular Mechanisms of Transcription Activation by Juvenile Hormone: A Critical Role for bHLH-PAS and Nuclear Receptor Proteins. INSECTS 2012; 3:324-38. [PMID: 26467963 PMCID: PMC4553631 DOI: 10.3390/insects3010324] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 03/15/2012] [Accepted: 03/16/2012] [Indexed: 11/16/2022]
Abstract
Juvenile hormone (JH) is responsible for controlling many biological processes. In several insect species JH has been implicated as a key regulator of developmental timing, preventing the premature onset of metamorphosis during larval growth periods. However, the molecular basis of JH action is not well-understood. In this review, we highlight recent advances which demonstrate the importance of transcription factors from the bHLH-PAS and nuclear receptor families in mediating the response to JH.
Collapse
Affiliation(s)
| | - Edward B Dubrovsky
- Department of Biology, Fordham University, Bronx, NY 10458, USA.
- Center for Cancer, Genetic Diseases, and Gene Regulation, Fordham University, Bronx, NY 10458, USA.
| |
Collapse
|
18
|
Largy E, Hamon F, Teulade-Fichou MP. A streptavidin paramagnetic-particle based competition assay for the evaluation of the optical selectivity of quadruplex nucleic acid fluorescent probes. Methods 2012; 57:129-37. [PMID: 22406492 DOI: 10.1016/j.ymeth.2012.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/18/2012] [Accepted: 02/20/2012] [Indexed: 01/29/2023] Open
Abstract
Although quadruplex nucleic acids are thought to be involved in many biological processes, they are massively overwhelmed by duplex DNA in the cell. Small molecules, able to probe quadruplex nucleic acids with high optical selectivity, could possibly achieve the visualization of these processes. The aim of the method described herein is to evaluate quickly the optical selectivity of quadruplex nucleic acid probes, in isothermal conditions, using widely available materials, small quantities of oligonucleotides and virtually any kind and quantity of biological competitor. The assay relies on the use of streptavidin-coated paramagnetic particles and biotinylated quadruplex forming oligonucleotides, allowing a quick and easy separation of the quadruplex target from the competitor. In the present study, two quadruplex nucleic acids (the DNA and RNA human telomeric repeats) have been used as targets while a duplex DNA oligonucleotide, total DNA, total RNA, another quadruplex nucleic acid and a protein have been used as competitors. The optical selectivity of various probes, displaying different photophysical properties and binding selectivities, has been successfully examined, allowing the identification of a best candidate for further cell microscopy experiments. This assay allows a quick and reliable assessment of the labeling properties of a quadruplex binder in cellular environment conditions. It is an interesting alternative to gel electrophoresis experiments since it is performed in solution, has a well-resolved separation system and allows easy quantifications.
Collapse
Affiliation(s)
- Eric Largy
- UMR 176 - Synthèse et Vectorisation de Biomolécules, Institut Curie, Bât. 110-112, Université Paris-Sud, 91405 Orsay, France.
| | | | | |
Collapse
|
19
|
Ben Saïd M, Ayedi L, Mnejja M, Hakim B, Khalfallah A, Charfeddine I, Khifagi C, Turki K, Ayadi H, Benzina Z, Ghorbel A, Castillo ID, Masmoudi S, Aifa MH. A novel missense mutation in the ESRRB gene causes DFNB35 hearing loss in a Tunisian family. Eur J Med Genet 2011; 54:e535-41. [PMID: 21802533 DOI: 10.1016/j.ejmg.2011.06.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 06/20/2011] [Indexed: 11/18/2022]
Abstract
Autosomal recessive non-syndromic hearing loss (ARNSHL) is a genetically heterogenous disorder with 41 genes so far identified. Among these genes, ESRRB whose mutations are responsible for DFNB35 hearing loss in Pakistani and Turkish families. This gene encodes the estrogen-related receptor beta. In this study, we report a novel mutation (p.Y305H) in the ESRRB gene in a Tunisian family with ARNSHL. This mutation was not detected in 100 healthy individuals. Molecular modeling showed that the p.Y305H mutation is likely to alter the conformation of the ligand binding-site by destabilizing the coactivator binding pocket. Interestingly, this ligand-binding domain of the ESRRB protein has been affected in 5 out of 6 mutations causing DFNB35 hearing loss. Using linkage and DHPLC analysis, no more mutations were detected in the ESRRB gene in other 127 Tunisian families with ARNSHL indicating that DFNB35 is most likely to be a rare type of ARNSHL in the Tunisian population.
Collapse
Affiliation(s)
- Mariem Ben Saïd
- Equipe Procédés de Criblages Moléculaires et Cellulaires, Laboratoire de Microorganismes et de Biomolécules, Centre de Biotechnologie de Sfax, Université de Sfax, Tunisia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Möcklinghoff S, Rose R, Carraz M, Visser A, Ottmann C, Brunsveld L. Synthesis and crystal structure of a phosphorylated estrogen receptor ligand binding domain. Chembiochem 2011; 11:2251-4. [PMID: 20922740 DOI: 10.1002/cbic.201000532] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sabine Möcklinghoff
- Department of Biomedical Engineering, Technische Universiteit Eindhoven, Eindhoven, NL
| | | | | | | | | | | |
Collapse
|
21
|
Christian CA, Glidewell-Kenney C, Jameson JL, Moenter SM. Classical estrogen receptor alpha signaling mediates negative and positive feedback on gonadotropin-releasing hormone neuron firing. Endocrinology 2008; 149:5328-34. [PMID: 18635656 PMCID: PMC2584581 DOI: 10.1210/en.2008-0520] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Accepted: 07/09/2008] [Indexed: 01/22/2023]
Abstract
During the female reproductive cycle, the neuroendocrine action of estradiol switches from negative feedback to positive feedback to initiate the preovulatory GnRH and subsequent LH surges. Estrogen receptor-alpha (ERalpha) is required for both estradiol negative and positive feedback regulation of LH. ERalpha may signal through estrogen response elements (EREs) in DNA and/or via ERE-independent pathways. Previously, a knock-in mutant allele (ERalpha-/AA) that selectively restores ERE-independent signaling onto the ERalpha-/- background was shown to confer partial negative but not positive estradiol feedback on serum LH. The current study investigated the roles of the ERE-dependent and ERE-independent ERalpha pathways for estradiol feedback at the level of GnRH neuron firing activity. The above ERalpha genetic models were crossed with GnRH-green fluorescent protein mice to enable identification of GnRH neurons in brain slices. Targeted extracellular recordings were used to monitor GnRH neuron firing activity using an ovariectomized, estradiol-treated mouse model that exhibits diurnal switches between negative and positive feedback. In wild-type mice, GnRH neuron firing decreased in response to estradiol during negative feedback and increased during positive feedback. In contrast, both positive and negative responses to estradiol were absent in GnRH neurons from ERalpha-/- and ERalpha-/AA mice. ERE-dependent signaling is thus required to increase GnRH neuron firing to generate a GnRH/LH surge. Furthermore, ERE-dependent and -independent ERalpha signaling pathways both appear necessary to mediate estradiol negative feedback on serum LH levels, suggesting central and pituitary estradiol feedback may use different combinations of ERalpha signaling pathways.
Collapse
Affiliation(s)
- Catherine A Christian
- Neuroscience Graduate Program, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
22
|
Zaman G, Jessop HL, Muzylak M, De Souza RL, Pitsillides AA, Price JS, Lanyon LL. Osteocytes use estrogen receptor alpha to respond to strain but their ERalpha content is regulated by estrogen. J Bone Miner Res 2006; 21:1297-306. [PMID: 16869728 DOI: 10.1359/jbmr.060504] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED The role of mechanical strain and estrogen status in regulating ERalpha levels in bone cells was studied in female rats. OVX is associated with decreased ERalpha protein expression/osteocyte, whereas habitual strain and artificial loading has only a small but positive effect, except on the ulna's medial surface, where artificial loading stimulates reversal of resorption to formation. INTRODUCTION Osteoporosis is the most widespread failure of bones' ability to match their architectural strength to their habitual load bearing. In men and women, the severity of bone loss is associated with bioavailability of estrogen. This association could result from the estrogen receptor (ER) involvement in bone cells' adaptive response to loading. MATERIALS AND METHODS In vivo semiquantitative analysis of the amount of ERalpha protein per osteocyte was performed in immuno-cytochemically stained sections from control and loaded rat ulna, as well as tibias of ovariectomy (OVX) and sham-operated female rats. In vitro, the effect of exogenous estrogen (10(-8) M) and mechanical strain (3400 microepsilon, 1 Hz, 600 cycles) on the expression of ERalpha mRNA levels was assessed in ROS 17/2.8 cells in monolayers using real-time PCR and ER promoter activity. ERalpha translocation in response to exogenous estrogen and mechanical strain was assessed in both ROS 17/2.8 and MLO-Y4 cells. RESULTS More than 90 percent of tibial osteocytes express ERalpha, the level/osteocyte being higher in cortical than cancellous bone. OVX is associated with decreased ERalpha protein expression/osteocyte, whereas in the ulna habitual strain and that caused by artificial loading had only a small but positive effect, except on the medial surface, where loading stimulates reversal of resorption to formation. In unstimulated osteocytes and osteoblasts in situ, and osteocyte-like and osteoblast-like cells in vitro, ERalpha is predominantly cytoplasmic. In vitro, both strain and estrogen stimulate transient ERalpha translocation to the nucleus and transient changes in ERalpha mRNA. Strain but not estrogen also induces discrete membrane localization of ERalpha. CONCLUSIONS Bone cells' responses to both strain and estrogen involve ERalpha, but only estrogen regulates its cellular concentration. This is consistent with the hypothesis that bone loss associated with estrogen deficiency is a consequence of reduction in ERalpha number/activity associated with lower estrogen concentration reducing the effectiveness of bone cells' anabolic response to strain.
Collapse
Affiliation(s)
- Gul Zaman
- Department of Basic Sciences, The Royal Veterinary College, University of London, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
23
|
Zhao C, Abrams J, Skafar DF. Targeted mutation of key residues at the start of helix 12 in the hERalpha ligand-binding domain identifies the role of hydrogen-bonding and hydrophobic interactions in the activity of the protein. J Steroid Biochem Mol Biol 2006; 98:1-11. [PMID: 16191480 DOI: 10.1016/j.jsbmb.2005.06.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Accepted: 06/30/2005] [Indexed: 11/21/2022]
Abstract
Estradiol (E(2)) and tamoxifen exert their effects through two members of the nuclear receptor superfamily, estrogen receptor (ER)-alpha and -beta. We want to identify the key interactions linking ligand-binding and activity of the ERalpha. Asp-351 and Leu-536 participate in hydrogen bond (Asp-351) and hydrophobic (Leu-536) interactions at the start of helix 12 in the ligand-binding domain (LBD) of the ERalpha. Mutations at each position alter ER activity, but we do not know which is more important. We mutated these residues in combination and individually and assessed the activity of the mutated ERs in the absence and presence of E(2) and 4-OHT on an ERE-driven and an AP-1-driven promoter, as well as their ability to interact with coregulators. On an ERE-driven promoter, the residue at position 351 determined whether E(2) stimulated or reduced the activity of the ER, as well as the level of activity in the presence of 4-OHT. Surprisingly, mutation of both residues generally did not produce cumulative deleterious effects, and they exerted counterbalancing effects on the basal activity on both promoters. Our results identify the contributions of specific interactions to the activity of the hERalpha, and support the concept that this region couples ligand-binding with ER activity.
Collapse
Affiliation(s)
- Changqing Zhao
- Department of Physiology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201-1928, USA
| | | | | |
Collapse
|
24
|
Hess-Wilson JK, Boldison J, Weaver KE, Knudsen KE. Xenoestrogen action in breast cancer: impact on ER-dependent transcription and mitogenesis. Breast Cancer Res Treat 2005; 96:279-92. [PMID: 16328721 DOI: 10.1007/s10549-005-9082-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2005] [Accepted: 09/16/2005] [Indexed: 11/26/2022]
Abstract
Several estrogen mimics (xenoestrogens) inappropriately activate the estrogen receptor (ER) in the absence of endogenous ligand. Given the importance of the ER in breast cancer growth and regulation, delineating the impact of these agents under conditions related to tumor treatment is of significant importance. We examined the effect of two prevalent xenoestrogens (bisphenol A and coumestrol) on ER activation and ER-dependent mitogenesis in breast cancer cells. We show that the ability of these agents to induce mitogenesis was restricted to conditions of estrogen depletion, and that these agents failed to cooperate with estradiol to induce MCF-7 breast cancer cell growth. These observations are consistent with the impact of each agent specifically on exogenous ER activation as monitored in HeLa cells, wherein the xenoestrogens activated the receptor in the absence of estradiol but failed to cooperate with estrogen. Tamoxifen blocked bisphenol A and coumestrol-mediated ER activation, indicating that exposure to these agents is unlikely to disrupt such therapeutic intervention. The response of tumor-derived ER alleles to these xenoestrogens was also examined. Although the xenoestrogens failed to alter ER-Y537S function, the ER-D351Y mutant demonstrated an enhanced response to bisphenol A. Moreover, tamoxifen enhanced the agonistic effects of xenoestrogens on ER-D351Y. Lastly, we examined the impact of ER co-activator overexpression on xenoestrogen response. Bisphenol A and coumestrol exhibited differential responses to co-activators with regard to ER activation. However, when using mitogenesis as an endpoint, these co-activators were insufficient to provide a significant growth advantage. Combined, these data demonstrate that bisphenol A and coumestrol can impact ER activity and ER-dependent proliferation in breast cancer cells, but the influence of these agents is restricted to conditions of estrogen depletion, selective mutation of the ER, and expression of specific co-activators.
Collapse
Affiliation(s)
- J K Hess-Wilson
- Department of Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267-0521, USA
| | | | | | | |
Collapse
|
25
|
Abstract
Endocrine therapy is the treatment of choice for patients with breast cancer expressing estrogen receptor (ER) and/or progesterone receptor. The efficacy of endocrine therapy is well established in the prevention, adjuvant and metastatic settings. However, either de novo or acquired resistance is frequently observed. Much effort has been made to elucidate the mechanisms of action underlying resistance to endocrine therapy in breast cancer, and several possible explanations have been suggested. Our previous studies have indicated that combined treatment with an antiestrogen, fulvestrant, and an inhibitor of the HER2 signaling pathway, trastuzumab, or an inhibitor of the HER1 signaling pathway, gefitinib, leads to an additive antitumor effect in breast cancer cells expressing ER and HER2 or HER1, respectively. It has also been suggested that the HER1 or HER2 signaling pathway is upregulated during the development of antiestrogen-resistant growth in breast cancer cells. These findings suggest that signal transduction inhibitors are effective for the treatment of antiestrogen-resistant breast cancer. A hypoxic microenvironment has been shown to promote malignant progression in cancer cells. Our previous study and others have suggested that hypoxia posttranscriptionally reduces ER expression and decreases sensitivity to hormonal agents in breast cancer cells. Our preliminary study has also shown that a hypoxic cytotoxin, tirapazamine, increases ER expression in breast cancer xenografts. Differential antitumor activity of tirapazamine on tumor cells under normoxic or hypoxic conditions may cause this phenomenon. These findings suggest that hypoxic cytotoxins may retard the development of endocrine resistance induced by hypoxia. Molecular mechanisms responsible for endocrine resistance in breast cancer are reviewed and possible therapeutic strategies against this resistance are discussed.
Collapse
Affiliation(s)
- Junichi Kurebayashi
- Department of Breast and Thyroid Surgery, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| |
Collapse
|
26
|
Kim H, Laing M, Muller W. c-Src-null mice exhibit defects in normal mammary gland development and ERalpha signaling. Oncogene 2005; 24:5629-36. [PMID: 16007215 DOI: 10.1038/sj.onc.1208718] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The c-Src tyrosine kinase has been implicated to play an integral role in modulating growth factor receptor, integrin and steroid receptor function. One class of steroid receptors that c-Src modulates is the estrogen receptor alpha (ERalpha). Although there is strong biochemical evidence supporting a role for c-Src in ERalpha signaling, the consequence of this association is unclear at the biological level. To explore the significance of c-Src in ERalpha signaling, we studied the development of various reproductive organs that are dependent on ERalpha in c-Src-deficient mice. We show that the loss of the c-Src tyrosine kinase correlates with defects in ductal development as well as in uterine and ovarian development. Genetic and biochemical analyses of c-Src-deficient mammary epithelial cells also revealed defects in the ability of mammary epithelial cells to activate a number of signaling pathways in response to exogenous estrogen stimulation. Taken together, these studies demonstrate that c-Src plays a role in ERalpha signaling in vivo.
Collapse
Affiliation(s)
- Harold Kim
- Department of Medical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada L8S 4K1
| | | | | |
Collapse
|
27
|
Cestac P, Doisneau-Sixou S, Favre G. Développement des inhibiteurs de farnésyl transférase comme agents anticancéreux. ANNALES PHARMACEUTIQUES FRANÇAISES 2005; 63:76-84. [PMID: 15803104 DOI: 10.1016/s0003-4509(05)82254-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ras proteins belong to the monomeric GTPases familly. They control cell growth, differentiation, proliferation, and survival. Ras mutations are frequently found in human cancers and play a fundamental role in tumorigenesis. Ras requires localization to the plasma membrane to exert its oncogenic effects. This subcelllular localization is dependent of protein farnesylation which is a post translational modification catalysed by the farnesyl transferase enzyme. Farnesyl transferase Inhibitors (FTI) were then designed ten to twelve years ago to inhibit ras processing and consequently the growth of ras mutated tumor. Preclinical data show that FTIs inhibit cell proliferation and survival in vitro and in vivo of a wide range of cancer cell lines, many of which contain wild type ras suggesting that mutated Ras is not the only target of the FTIs effects. Four FTIs went then through clinical trials and three of then are still developed in the clinic. Phase I et II clinical trials confirmed a relevant antitumor activity and a low toxicity. Phase III clinical trials are currently undergoing for both solid and hematologic tumors. The expected results should allow to define the position of FTIs as anticancer drugs, particularly in combination with conventional chemotherapy, hormone therapy, radiotherapy or any other new targeted compound.
Collapse
Affiliation(s)
- Ph Cestac
- Inserm U563, Département innovation thérapeutique et oncologie moléculaire, F31052 Toulouse, France
| | | | | |
Collapse
|
28
|
Wang MM, Traystman RJ, Hurn PD, Liu T. Non-classical regulation of estrogen receptor-alpha by ICI182,780. J Steroid Biochem Mol Biol 2004; 92:51-62. [PMID: 15544930 DOI: 10.1016/j.jsbmb.2004.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Accepted: 06/14/2004] [Indexed: 11/26/2022]
Abstract
Estrogen receptor-alpha (ER alpha) regulates transcription through a number of molecular mechanisms. Two mechanisms by which ER alpha acts directly in the nucleus have emerged: (1) in classical ER alpha action, estrogen-bound receptor binds estrogen response elements (ERE) and regulates promoters by recruiting coactivators or corepressors to DNA; (2) non-classical action is not dependent on ER alpha binding to EREs; its mechanism is not as clearly defined as classical action. In many instances, non-classical action is mediated by tethering of ER alpha to other DNA-binding proteins, facilitating recruitment of coregulators to transcription regulatory sequences. In some cell types, non-classical stimulation can be enhanced by antagonists and repressed by agonists of ER alpha. Here, we show that non-classical action of ER alpha in 293 cells occurs in a wide range of enhancers and enhancer binding proteins. ER alpha stimulates AP-1 elements, cyclic AMP response elements (CRE), and serum response elements (SRE) in the presence of the antiestrogen ICI182,780. Further, in the presence of ICI182,780, ER alpha stimulates activation domains of Jun, ATF-2, Elk, and CRE-binding protein (CREB). Non-classical ER alpha regulation described here does not appear to be sensitive to point mutations which affect classical and tethered ER alpha action; moreover, in our experiments, non-classical action is uniquely sensitive to nuclear transport inhibition by leptomycin B. Because ICI182,780 appears to affect multiple and diverse transcriptional systems, our results are likely explained by ER alpha-dependent modulation of common components of the transcriptional machinery and may not be completely explained by tethering of ER alpha to specific transcription factors.
Collapse
Affiliation(s)
- Michael M Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-0622, USA.
| | | | | | | |
Collapse
|
29
|
Bardin A, Boulle N, Lazennec G, Vignon F, Pujol P. Loss of ERbeta expression as a common step in estrogen-dependent tumor progression. Endocr Relat Cancer 2004; 11:537-51. [PMID: 15369453 PMCID: PMC2072930 DOI: 10.1677/erc.1.00800] [Citation(s) in RCA: 314] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The characterization of estrogen receptor beta (ERbeta) brought new insight into the mechanisms underlying estrogen signaling. Estrogen induction of cell proliferation is a crucial step in carcinogenesis of gynecologic target tissues, and the mitogenic effects of estrogen in these tissues (such as breast, endometrium and ovary) are well documented both in vitro and in vivo. There is also an emerging body of evidence that colon and prostate cancer growth is influenced by estrogens. In all of these tissues, most studies have shown decreased ERbeta expression in cancer as compared with benign tumors or normal tissues, whereas ERalpha expression persists. The loss of ERbeta expression in cancer cells could reflect tumor cell dedifferentiation but may also represent a critical stage in estrogen-dependent tumor progression. Modulation of the expression of ERalpha target genes by ERbeta or ERbeta-specific gene induction could explain that ERbeta has a differential effect on proliferation as compared with ERalpha. ERbeta may exert a protective effect and thus constitute a new target for hormone therapy, such as ligand specific activation. The potential distinct roles of ERalpha and ERbeta expression in carcinogenesis, as suggested by experimental and clinical data, are discussed in this review.
Collapse
Affiliation(s)
- Allison Bardin
- Endocrinologie moléculaire et cellulaire des cancers
INSERM : U540 INSERM : U540Université Montpellier I60 rue de Navacelles
34090 Montpellier,FR
| | - Nathalie Boulle
- Endocrinologie moléculaire et cellulaire des cancers
INSERM : U540 INSERM : U540Université Montpellier I60 rue de Navacelles
34090 Montpellier,FR
- Laboratoire de Biologie Cellulaire et
Hormonale
CHRU MontpellierHôpital Arnaud de Villeneuve,FR
| | - Gwendal Lazennec
- Endocrinologie moléculaire et cellulaire des cancers
INSERM : U540 INSERM : U540Université Montpellier I60 rue de Navacelles
34090 Montpellier,FR
- * Correspondence should be adressed to: Gwendal Lazennec
| | - Françoise Vignon
- Endocrinologie moléculaire et cellulaire des cancers
INSERM : U540 INSERM : U540Université Montpellier I60 rue de Navacelles
34090 Montpellier,FR
| | - Pascal Pujol
- Endocrinologie moléculaire et cellulaire des cancers
INSERM : U540 INSERM : U540Université Montpellier I60 rue de Navacelles
34090 Montpellier,FR
- Laboratoire de Biologie Cellulaire et
Hormonale
CHRU MontpellierHôpital Arnaud de Villeneuve,FR
- * Correspondence should be adressed to: Pascal Pujol
| |
Collapse
|
30
|
Barletta F, Wong CW, McNally C, Komm BS, Katzenellenbogen B, Cheskis BJ. Characterization of the Interactions of Estrogen Receptor and MNAR in the Activation of cSrc. Mol Endocrinol 2004; 18:1096-108. [PMID: 14963108 DOI: 10.1210/me.2003-0335] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
AbstractIn this study, we have evaluated the molecular mechanism of Src activation after its interaction with estrogen receptor α (ERα) and a newly identified scaffold protein, called MNAR (modulator of nongenomic activity of ER). Under basal condition, Src enzymatic activity is inhibited by intramolecular interactions. The enzyme can be activated by interaction between the SH2 domain of Src and phosphotyrosine-containing sequences and/or by interaction between the SH3 domain of Src and proteins containing PXXP motifs. Mutational analysis and functional evaluation of MNAR and the use of ERα and cSrc mutants revealed that MNAR interacts with Src’s SH3 domain via its N-terminal PXXP motif. Mutation of this motif abolished both the MNAR-induced activation of Src and the stimulation of ER transcriptional activity. ER interacts with Src’s SH2 domain using phosphotyrosine 537, and this complex was further stabilized by MNAR-ER interaction. Mapping studies reveal that both the A/B domain and Y537 of ERα are required for MNAR-induced activation of ER transcriptional activity. The region responsible for MNAR interaction with ER maps to two N-terminal LXXLL motifs of MNAR. Mutation of these motifs prevented ER-MNAR complex formation and eliminated activation of the Src/MAPK pathway. These data explicate how the coordinate interactions between MNAR, ER, and Src lead to Src activation. Our findings also demonstrate that MNAR is a scaffold protein that mediates ER-Src interaction and plays an important role in the integration of ER action in Src-mediated signaling.
Collapse
Affiliation(s)
- Frank Barletta
- Department of Women's Health and Bone Research, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426, USA
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Estrogens work along with genetic changes to promote the development and growth of breast cancers. Because estrogenic hormones act via the estrogen receptors (ERs), ER-alpha and ER-beta, and the ER is present in more than half of breast tumors, this receptor has been the most widely targeted protein in breast cancer therapy. The presence of the ER in breast tumors predicts improved disease-free survival and response to selective ER modulators (SERMs), such as tamoxifen, or other forms of endocrine therapy. Suppression of ER activity by SERMs has proven to be a great benefit in the treatment of breast cancers and also in the prevention of breast cancer in women at high risk for the disease. The Study of Tamoxifen and Raloxifene trial comparing tamoxifen versus raloxifene effectiveness in breast cancer prevention is currently under way. To understand the balance of beneficial and undesirable effects of SERMs and to optimize their effectiveness, current investigations seek to characterize the genes activated or suppressed by these agents. Elucidation of the gene networks and cell signaling pathways under estrogen and SERM regulation and a clearer definition of the respective roles of ER-alpha and ER-beta and their coregulators in the actions of selective ER ligands, should enable the identification of new gene targets for therapeutic intervention and the development of novel drugs for the optimal treatment and prevention of breast cancer.
Collapse
Affiliation(s)
- Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois and College of Medicine, Urbana, IL, USA
| | | |
Collapse
|
32
|
Matsuda S, Harries JC, Viskaduraki M, Troke PJF, Kindle KB, Ryan C, Heery DM. A Conserved alpha-helical motif mediates the binding of diverse nuclear proteins to the SRC1 interaction domain of CBP. J Biol Chem 2004; 279:14055-64. [PMID: 14722092 DOI: 10.1074/jbc.m310188200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
CREB-binding protein (CBP) and p300 contain modular domains that mediate protein-protein interactions with a wide variety of nuclear factors. A C-terminal domain of CBP (referred to as the SID) is responsible for interaction with the alpha-helical AD1 domain of p160 coactivators such as the steroid receptor coactivator (SRC1), and also other transcriptional regulators such as E1A, Ets-2, IRF3, and p53. Here we show that the pointed (PNT) domain of Ets-2 mediates its interaction with the CBP SID, and describe the effects of mutations in the SID on binding of Ets-2, E1A, and SRC1. In vitro binding studies indicate that SRC1, Ets-2 and E1A display mutually exclusive binding to the CBP SID. Consistent with this, we observed negative cross-talk between ERalpha/SRC1, Ets-2, and E1A proteins in reporter assays in transiently transfected cells. Transcriptional inhibition of Ets-2 or GAL4-AD1 activity by E1A was rescued by co-transfection with a CBP expression plasmid, consistent with the hypothesis that the observed inhibition was due to competition for CBP in vivo. Sequence comparisons revealed that SID-binding proteins contain a leucine-rich motif similar to the alpha-helix Aalpha1 of the SRC1 AD1 domain. Deletion mutants of E1A and Ets-2 lacking the conserved motif were unable to bind the CBP SID. Moreover, a peptide corresponding to this sequence competed the binding of full-length SRC1, Ets-2, and E1A proteins to the CBP SID. Thus, a leucine-rich amphipathic alpha-helix mediates mutually exclusive interactions of functionally diverse nuclear proteins with CBP.
Collapse
Affiliation(s)
- Sachiko Matsuda
- Department of Biochemistry, University of Leicester, University Road Leicester LE1 7RH, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
33
|
Buluwela L, Constantinidou D, Pike J, Ali S. Estrogen receptors and anti-estrogen therapies. Cancer Treat Res 2004; 119:271-92. [PMID: 15164882 DOI: 10.1007/1-4020-7847-1_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Affiliation(s)
- Lakjaya Buluwela
- Department of Cancer Medicine, Imperial College London, United Kingdom
| | | | | | | |
Collapse
|
34
|
Kurebayashi J. Endocrine-resistant breast cancer: underlying mechanisms and strategies for overcoming resistance. Breast Cancer 2003; 10:112-9. [PMID: 12736563 DOI: 10.1007/bf02967635] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Estrogen plays important roles in the development and progression of breast cancer. However, one-third of breast cancers fail to respond to endocrine therapy and most endocrine-responsive breast cancers subsequently become resistant to endocrine therapy. A tremendous effort has been made to elucidate the mechanisms responsible for the development of endocrine-resistance in breast cancer. Since the main target molecule of estrogen in breast cancer is estrogen receptor (ER)-alpha, most studies have focused on investigating quantitative and qualitative changes in ER-alpha in endocrine-resistant breast cancer. Breast cancers expressing no ER-alpha fail to respond to endocrine therapy. Some breast cancers expressing ER-alpha also fail to respond to endocrine therapy and most breast cancers with acquired endocrine resistance retain ER-alpha expression, which suggests that the disappearance of ER-alpha in breast cancer cells is not a common cause of resistance to endocrine therapy. Recent molecular biological studies have shown evidence that qualitative and functional changes, such as gene mutations and phosphorylation of ER-alpha, cause endocrine resistance in breast cancer. In addition, it has been suggested that endocrine resistance could be induced by epigenetic changes, such as hypoxia, in breast cancer tissues. Understanding the precise mechanisms that underlie endocrine resistance may enable clinicians to develop new strategies for retarding or overcoming endocrine resistance in breast cancer.
Collapse
Affiliation(s)
- Junichi Kurebayashi
- Department of Breast and Thyroid Surgery, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| |
Collapse
|
35
|
Chen D, Lucey MJ, Phoenix F, Lopez-Garcia J, Hart SM, Losson R, Buluwela L, Coombes RC, Chambon P, Schär P, Ali S. T:G mismatch-specific thymine-DNA glycosylase potentiates transcription of estrogen-regulated genes through direct interaction with estrogen receptor alpha. J Biol Chem 2003; 278:38586-92. [PMID: 12874288 DOI: 10.1074/jbc.m304286200] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nuclear receptors (NR) classically regulate gene expression by stimulating transcription upon binding to their cognate ligands. It is now well established that NR-mediated transcriptional activation requires the recruitment of coregulator complexes, which facilitate recruitment of the basal transcription machinery through direct interactions with the basal transcription machinery and/or through chromatin remodeling. However, a number of recently described NR coactivators have been implicated in cross-talk with other nuclear processes including RNA splicing and DNA repair. T:G mismatch-specific thymine DNA glycosylase (TDG) is required for base excision repair of deaminated methylcytosine. Here we show that TDG is a coactivator for estrogen receptor alpha (ERalpha). We demonstrate that TDG interacts with ERalpha in vitro and in vivo and suggest a separate role for TDG to its established role in DNA repair. We show that this involves helix 12 of ERalpha. The region of interaction in TDG is mapped to a putative alpha-helical motif containing a motif distinct from but similar to the LXXLL motif that mediates interaction with NR. Together with recent reports linking TFIIH in regulating NR function, our findings provide new data to further support an important link between DNA repair proteins and nuclear receptor function.
Collapse
Affiliation(s)
- Dongsheng Chen
- Department of Cancer Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ray S, Rastogi R, Kumar A. Current status of estrogen receptors. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 59:201-32. [PMID: 12458968 DOI: 10.1007/978-3-0348-8171-5_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Increasing knowledge on structure and function of estrogen receptors is providing information on the mechanism of action of estrogen agonists, as well as antagonists, and in understanding their tissue-selective action. However, there are still many factors associated with estrogen response which are poorly understood. Therefore, the task of designing a tissue-selective estrogen for use as a pharmaceutical in estrogen-dependent disorders remains an uncertain game. This review provides information on the current status of estrogen receptors for a better understanding.
Collapse
Affiliation(s)
- Suprabhat Ray
- Medicinal Chemistry Division, Central Drug Research Institute, Lucknow 226001, India.
| | | | | |
Collapse
|
37
|
Abstract
Estrogen receptor alpha (ERalpha) is phosphorylated on multiple amino acid residues. For example, in response to estradiol binding, human ERalpha is predominately phosphorylated on Ser-118 and to a lesser extent on Ser-104 and Ser-106. In response to activation of the mitogen-activated protein kinase pathway, phosphorylation occurs on Ser-118 and Ser-167. These serine residues are all located within the activation function 1 region of the N-terminal domain of ERalpha. In contrast, activation of protein kinase A increases the phosphorylation of Ser-236, which is located in the DNA-binding domain. The in vivo phosphorylation status of Tyr-537, located in the ligand-binding domain, remains controversial. In this review, I present evidence that these phosphorylations occur, and identify the kinases thought to be responsible. Additionally, the functional importance of ERalpha phosphorylation is discussed.
Collapse
Affiliation(s)
- Deborah A Lannigan
- Center for Cell Signaling, Health Sciences Center, University of Virginia, Hospital West, Room 7041, Box 800577, Charlottesville, VA 22908-0577, USA.
| |
Collapse
|
38
|
Abstract
Nuclear receptors (NRs) comprise a family of 49 members that share a common structural organization and act as ligand-inducible transcription factors with major (patho)physiological impact. For some NRs (“orphan receptors”), cognate ligands have not yet been identified or may not exist. The principles of DNA recognition and ligand binding are well understood from both biochemical and crystal structure analyses. The 3D structures of several DNA-binding domains (DBDs),in complexes with a variety of cognate response elements, and multiple ligand-binding domains (LBDs), in the absence (apoLBD)and presence (holoLBD) of agonist, have been established and reveal canonical structural organization. Agonist binding induces a structural transition in the LBD whose most striking feature is the relocation of helix H12, which is required for establishing a coactivator complex, through interaction with members of the p160 family (SRC1, TIF2, AIB1) and/or the TRAP/DRIP complex. The p160-dependent coactivator complex is a multiprotein complex that comprises histone acetyltransferases (HATs), such as CBP,methyltransferases, such as CARM1, and other enzymes (SUMO ligase,etc.). The agonist-dependent recruitment of the HAT complex results in chromatin modification in the environment of the target gene promoters, which is requisite to, or may in some cases be sufficient for, transcription activation. In the absence of ligands, or in the presence of some antagonists, certain NRs are bound to distinct multiprotein complexes through the interaction with corepressors, such as NCoR and SMRT. Corepressor complexes comprise histone deacetylases (HDACs) that have the capacity to condense chromatin over target gene promoters. Ligands have been designed that selectively modulate the interaction between NRs and their coregulators. Both HATs and HDACs can also modify the acetylation status of nonhistone proteins, but the significance in the context of NR signaling is unclear. NRs communicate with other intracellular signaling pathways on a mutual basis, and their functionality may be altered, positively or negatively, by post-translational modification. The majority of NRs act as retinoid X receptor (RXR) heterodimers in which RXR cannot a priori respond autonomously to its cognate ligand to activate target gene transcription. This RXR subordination allows signaling pathway identity for the RXR partner. The corresponding mechanism is understood and reveals cell and NR selectivity, indicating that RXR can, under certain conditions, act autonomously. NRs are regulators of cell life and death,and NR malfunction can be at the basis of both disease and therapy, as is impressively documented in the case of acute promyelocytic leukemia. Recently, several pathways have been uncovered that link NR action with cell proliferation and apoptosis.
Collapse
|
39
|
Abstract
There is still extensive disparity in our understanding of how estrogens exert their actions, particularly in non-reproductive tissues such as the skin. Although it has been recognized for some time that estrogens have significant effects on many aspects of skin physiology and pathophysiology, studies on estrogen action in skin have been limited. However, estrogens clearly have an important function in many components of human skin including the epidermis, dermis, vasculature, hair follicle and the sebaceous, eccrine and apocrine glands, having significant roles in skin aging, pigmentation, hair growth, sebum production and skin cancer. The recent discovery of a second intracellular estrogen receptor (ERbeta) with different cell-specific roles to the classic estrogen receptor (ERalpha), and the identification of cell surface estrogen receptors, has provided further challenges to understanding the mechanism of estrogen action. It is now time to readdress many of the outstanding questions regarding the role of estrogens in skin and improve our understanding of the physiology and interaction of steroid hormones and their receptors in human skin. Not only will this lead to a better understanding of estrogen action, but may also provide a basis for further interventions in pathological processes that involve dysregulation of estrogen action.
Collapse
Affiliation(s)
- M J Thornton
- Department of Biomedical Sciences, University of Bradford, Bradford, West Yorkshire, UK.
| |
Collapse
|
40
|
Björnström L, Sjöberg M. Signal transducers and activators of transcription as downstream targets of nongenomic estrogen receptor actions. Mol Endocrinol 2002; 16:2202-14. [PMID: 12351686 DOI: 10.1210/me.2002-0072] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
17Beta-estradiol-activated estrogen receptor alpha (ERalpha) and beta (ERbeta) are able to induce transcriptional activation of signal transducer and activator of transcription (Stat)-regulated promoters via cytoplasmic signal transduction pathways. Stat5 and Stat3 are required for promoter induction, which correlates with cytoplasmic sublocalization of ERs and is independent of intact coactivator binding sites and DNA-binding domains. In endothelial cells, Stat5 and Stat3 are rapidly phosphorylated on both tyrosine and serine residues in response to 17beta-estradiol, and nuclear translocation is subsequently induced. 17Beta-estradiol-induced transactivation of a Stat-regulated promoter requires at least three different signal transduction pathways, including MAPK, Src-kinase, and phosphatidylinositol-3-kinase activities. In conclusion, this work identifies a novel pathway involving an agonist-bound ER-activated phosphorylation cascade, resulting in nuclear transcriptional activation of target transcription factors. These findings reveal novel targets for the development of drugs that modulate a nongenomic-to-genomic ER-dependent mechanism.
Collapse
Affiliation(s)
- Linda Björnström
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | | |
Collapse
|
41
|
Ostberg T, Bertilsson G, Jendeberg L, Berkenstam A, Uppenberg J. Identification of residues in the PXR ligand binding domain critical for species specific and constitutive activation. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:4896-904. [PMID: 12354121 DOI: 10.1046/j.1432-1033.2002.03207.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The cytochrome P450 family of enzymes has long been known to metabolize a wide range of compounds, including many of today's most common drugs. A novel nuclear receptor called PXR has been established as an activator of several of the cytochrome P450 genes, including CYP3A4. This enzyme is believed to account for the metabolism of more than 50% of all prescription drugs. PXR is therefore used as a negative selector target and discriminatory filter in preclinical drug development. In this paper we describe the design, construction and characterization by transient transfection of mutant receptors of the human and mouse PXR ligand binding domains. By modeling the human PXR ligand binding domain we have identified and mutated two polar residues in the putative ligand binding pocket which differ between the human and the mouse receptor. The first residue (Q285 in human/I282 in mouse) was mutated between the two species with the corresponding amino acids. These mutants showed that this residue is important for the species specific activation of PXR by the ligand pregnenolone-16alpha-carbonitrile (PCN), while having a less pronounced role in receptor activation by rifampicin. The second residue to be mutated (H407 in human/Q404 in mouse) unexpectedly proved to be important for the basal level of activation of PXR. The H407A mutant of the human receptor showed a high level of constitutive activity, while the Q404H mutant of the mouse receptor demonstrated a sharply decreased basal activity compared to wild-type.
Collapse
Affiliation(s)
- Tove Ostberg
- Department of Cell and Molecular Biology, Medical Nobel Institute, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
42
|
McDonnell DP, Wijayaratne A, Chang CY, Norris JD. Elucidation of the molecular mechanism of action of selective estrogen receptor modulators. Am J Cardiol 2002; 90:35F-43F. [PMID: 12106639 DOI: 10.1016/s0002-9149(01)02221-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The term selective estrogen receptor modulator (SERM) describes a group of pharmaceuticals that manifest estrogen receptor (ER) agonist activity in some tissues but opposes estrogen action in others. Although the name describing this class of drugs is new, the concept is not, as compounds exhibiting tissue-selective ER agonist/antagonist properties have been available for nearly 40 years. What is new is the idea that it may be possible to capitalize on the paradoxical activities of SERMs and develop them as target organ-selective ER agonists for the treatment of osteoporosis and other estrogenopathies. This realization has provided the impetus for research in this area, the progress of which is described in this review.
Collapse
Affiliation(s)
- Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | |
Collapse
|
43
|
Zhong L, Skafar DF. Mutations of tyrosine 537 in the human estrogen receptor-alpha selectively alter the receptor's affinity for estradiol and the kinetics of the interaction. Biochemistry 2002; 41:4209-17. [PMID: 11914066 DOI: 10.1021/bi0121095] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mutation of tyrosine 537 (Y537) of the human estrogen receptor-alpha (hERalpha) produces receptors having a range of constitutive activity, which suggests that this residue modulates the conformational changes of the receptor. We investigated the effect of several mutations at this position, to phenylalanine (Y537F), to serine (Y537S), and to glutamic acid (Y537E), on the hormone-binding properties of the receptor. The affinities of the wt, the Y537F mutant, and the Y537S mutant for estradiol were similar: K(a) = 2.2 +/- 0.2, 3.9 +/- 0.5, and 2.8 +/- 0.4 nM(-1), respectively. By contrast, the affinity of the Y537E mutant for estradiol was reduced 10-fold, K(a) = 0.2 +/- 0.1 nM(-1). All proteins bound [(3)H]estradiol with a positive cooperative mechanism (n(H) = 1.7-1.9), indicating they can form dimers. The wt receptor and the Y537S and Y537E mutants exhibited biphasic dissociation kinetics, which is also indicative of dimerization. Surprisingly, the half-lives of the slow component of the wt and the Y537E mutant were indistinguishable, 118 +/- 3.4 and 122 +/- 4.5 min, respectively, even though the affinity of the Y537E mutant for hormone was reduced 10-fold. The half-life of the slow component of the Y537S mutant was reduced to 96.5 +/- 3.8 min. Molecular models were constructed and compared to identify changes in the structure that correlate with the observed effects on hormone binding. Local alterations in hydrogen bonding, the position of side chains, and the position of the peptide backbone were observed. Taken together, these results show that mutations at Y537 selectively alter the affinity and kinetics of hormone binding to the receptor, and are consistent with the idea that the estradiol-estrogen receptor interaction can follow more than one pathway.
Collapse
Affiliation(s)
- L Zhong
- Department of Physiology and Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | |
Collapse
|
44
|
Maeda M. The conserved residues of the ligand-binding domains of steroid receptors are located in the core of the molecules. J Mol Graph Model 2002; 19:543-51, 601-6. [PMID: 11552682 DOI: 10.1016/s1093-3263(01)00087-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The relationship between conserved residues and biochemical functions of steroid receptors was investigated. Pairwise three-dimensional (3D) alignment of the ligand-binding domains of the human estrogen (1A52) and progesterone (1A28) receptors revealed two conserved domains; Asn313-Ser456 and Gln471-Lys531 (numbering reflects the sequence in the human estrogen receptor). Alignment of the protein sequences of 39 steroid receptors revealed 36 highly conserved residues (i.e., the residues commonly found in more than 80% of sequences aligned). They were distributed throughout the sequences but formed a contiguous 3D structure. Most of these highly conserved residues were buried in the ligand-binding domain, but several residues were exposed on the surface. The well-known functions commonly associated with the ligand-binding domain of steroid receptors are ligand binding, HSP90 binding, transcriptional activation and dimerization. The relationship between the residues and these functions were checked. To determine the residues involved in dimerization, the differences between the solvent accessibilities of the monomeric and dimeric forms were calculated. These results revealed 32 residues of 1A52 and 15 residues of 1A28 potentially involved in dimerization. Their distribution areas do not overlap greatly. Comparing these putative dimerization sites with highly conserved residues, many of the exposed conserved residues were observed on the side of the domain opposite are the dimerization sites. Some highly conserved residues are located in a steroid-binding site and in transcriptional activation domain. However, few of them were observed in the HSP90 binding site. These results indicate that the core structure made by most of the highly conserved residues among the ligand-binding domains of steroid receptors is important. These conserved residues may be essential for conformational change in the ligand-binding domain from its inactive to active form.
Collapse
Affiliation(s)
- M Maeda
- Biochemistry Department, National Institute of Agrobiological Sciences, Kannondai 2-1-2, Tsukuba, Ibaraki 305-8602, Japan.
| |
Collapse
|
45
|
Koide A, Abbatiello S, Rothgery L, Koide S. Probing protein conformational changes in living cells by using designer binding proteins: application to the estrogen receptor. Proc Natl Acad Sci U S A 2002; 99:1253-8. [PMID: 11818562 PMCID: PMC122176 DOI: 10.1073/pnas.032665299] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A challenge in understanding the mechanism of protein function in biology is to establish the correlation between functional form in the intracellular environment and high-resolution structures obtained with in vitro techniques. Here we present a strategy to probe conformational changes of proteins inside cells. Our method involves: (i) engineering binding proteins to different conformations of a target protein, and (ii) using them to sense changes in the surface property of the target in cells. We probed ligand-induced conformational changes of the estrogen receptor alpha (ER alpha) ligand-binding domain (LBD). By using yeast two-hybrid techniques, we first performed combinatorial library screening of "monobodies" (small antibody mimics using the scaffold of a fibronectin type III domain) for clones that bind to ER alpha and then characterized their interactions with ER alpha in the nucleus, the native environment of ER alpha, in the presence of various ligands. A library using a highly flexible loop yielded monobodies that specifically recognize a particular ligand complex of ER alpha, and the pattern of monobody specificity was consistent with the structural differences found in known crystal structures of ER alpha-LBD. A more restrained loop library yielded clones that bind both agonist- and antagonist-bound ER alpha. Furthermore, we found that a deletion of the ER alpha F domain that is C-terminally adjacent to the LBD increased the crossreactivity of monobodies to the apo-ER alpha-LBD, suggesting a dynamic nature of the ER alpha-LBD conformation and a role of the F domain in restraining the LBD in an inactive conformation.
Collapse
Affiliation(s)
- Akiko Koide
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
46
|
Abstract
Deaths from breast cancer have fallen markedly over the past decade due, in part, to the use of endocrine agents that reduce the levels of circulating oestrogens or compete with oestrogen for binding to its receptor. However, many breast tumours either fail to respond or become resistant to endocrine therapies. By understanding the mechanisms that underlie this resistance, we might be able to develop strategies for overcoming or bypassing it.
Collapse
Affiliation(s)
- Simak Ali
- Department of Cancer Medicine and Cancer Research Campaign Laboratories, Faculty of Medicine, Imperial College of Science, Technology & Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK.
| | | |
Collapse
|
47
|
Walters MR, Dutertre M, Smith CL. SKF-82958 is a subtype-selective estrogen receptor-alpha (ERalpha ) agonist that induces functional interactions between ERalpha and AP-1. J Biol Chem 2002; 277:1669-79. [PMID: 11700319 DOI: 10.1074/jbc.m109320200] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The transcriptional activity of estrogen receptors (ERs) can be regulated by ligands as well as agents such as dopamine, which stimulate intracellular signaling pathways able to communicate with these receptors. We examined the ability of SKF-82958 (SKF), a previously characterized full dopamine D1 receptor agonist, to stimulate the transcriptional activity of ERalpha and ERbeta. Treatment of HeLa cells with SKF-82958 stimulated robust ERalpha-dependent transcription from an estrogen-response element-E1b-CAT reporter in the absence of estrogen, and this was accompanied by increased receptor phosphorylation. However, induction of ERbeta-directed gene expression under the same conditions was negligible. In our cell model, SKF treatment did not elevate cAMP levels nor enhance transcription from a cAMP-response element-linked reporter. Control studies revealed that SKF-82958, but not dopamine, competes with 17beta-estradiol for binding to ERalpha or ERbeta with comparable relative binding affinities. Therefore, SKF-82958 is an ERalpha-selective agonist. Transcriptional activation of ERalpha by SKF was more potent than expected from its relative binding activity, and further examination revealed that this synthetic compound induced expression of an AP-1 target gene in a tetradecanoylphorbol-13-acetate-response element (TRE)-dependent manner. A putative TRE site upstream of the estrogen-response element and the amino-terminal domain of the receptor contributed to, but were not required for, SKF-induced expression of an ERalpha-dependent reporter gene. Overexpression of the AP-1 protein c-Jun, but not c-Fos, strongly enhanced SKF-induced ERalpha target gene expression but only when the TRE was present. These studies provide information on the ability of a ligand that weakly stimulates ERalpha to yield strong stimulation of ERalpha-dependent gene expression through cross-talk with other intracellular signaling pathways producing a robust combinatorial response within the cell.
Collapse
Affiliation(s)
- Marian R Walters
- Department of Physiology, Tulane Medical School, New Orleans, Louisiana 70112, USA
| | | | | |
Collapse
|
48
|
McDonnell DP, Chang CY, Norris JD. Capitalizing on the complexities of estrogen receptor pharmacology in the quest for the perfect SERM. Ann N Y Acad Sci 2001; 949:16-35. [PMID: 11795348 DOI: 10.1111/j.1749-6632.2001.tb03999.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The term Selective Estrogen Receptor Modulators (SERMs) has been used of late to describe a group of pharmaceuticals that manifest estrogen receptor (ER) agonist activity in some tissues, but that oppose estrogen action in others. Whereas the name describing this class of drugs is new, the concept is not. Indeed, compounds exhibiting tissue-selective ER agonist/antagonist properties have been around for nearly 40 years. What is new is the idea that it may be possible to capitalize on the paradoxical activities of these drugs and develop them as treatments for estrogenopathies where it is desirable to direct therapy to a specific estrogen-responsive target organ. This realization has provided the impetus for research in this area and has pushed the development and clinical use of this class of drugs. The objective of this review is to describe how the medical need for SERMs arose and how recent studies of the mechanism of action of the currently available drugs are paving the way for the development of novel drugs with improved selectivity.
Collapse
Affiliation(s)
- D P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
49
|
Jetten AM, Kurebayashi S, Ueda E. The ROR nuclear orphan receptor subfamily: critical regulators of multiple biological processes. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2001; 69:205-47. [PMID: 11550795 DOI: 10.1016/s0079-6603(01)69048-2] [Citation(s) in RCA: 182] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The nuclear receptor superfamily, a group of structurally related, ligand-dependent transcription factors, includes a large number of orphan receptors for which no ligand has yet been identified. These proteins function as key regulators of many physiological processes that occur during embryonic development and in the adult. The retinoid-related orphan receptors (RORs) alpha, beta, and gamma comprise one nuclear orphan receptor gene subfamily. RORs exhibit a modular structure that is characteristic for nuclear receptors; the DNA-binding domain is highly conserved and the ligand-binding domain is moderately conserved among RORs. By a combination of alternative promoter usage and exon splicing, each ROR gene generates several isoforms that differ only in their amino terminus. RORs bind as monomers to specific ROR response elements (ROREs) consisting of the consensus core motif AGGTCA preceded by a 5-bp A/T-rich sequence. RORE-dependent transcriptional activation by RORs is cell type-specific and mediated through interactions with nuclear cofactors. RORs have been shown to interact with certain corepressors as well as coactivators, suggesting that RORs are not constitutively active but that their activity is under some regulatory control. RORs likely can assume at least two different conformations: a repressive state, which allows interaction with corepressor complexes, and an active state, which promotes binding of coactivator complexes. Whether the transition between these two states is regulated by ligand binding and/or by phosphorylation remains to be determined. Ca2+/calmodulin-dependent kinase IV (CaMKIV) can dramatically enhance ROR-mediated transcriptional activation. This stimulation involves CaMKIV-mediated phosphorylation not of RORs, but likely of specific nuclear cofactors that interact with RORs. RORalpha is widely expressed. In the cerebellum, its expression is limited to the Purkinje cells. RORalpha-/- mice and the natural RORalpha-deficient staggerer mice exhibit severe cerebellar ataxia due to a defect in Purkinje cell development. In addition, these mice have thin long bones, suggesting a role for RORalpha in bone metabolism, and develop severe atherosclerosis when placed on a high-fat diet. Expression of RORbeta is very restricted. RORbeta is highly expressed in different parts of the neurophotoendocrine system, the pineal gland, the retina, and suprachiasmatic nuclei, suggesting a role in the control of circadian rhythm. This is supported by observations showing alterations in circadian behavior in RORbeta-/- mice. RORgamma, which is most highly expressed in the thymus, plays an important role in thymopoiesis. Thymocytes from RORgamma-/- mice undergo accelerated apoptosis. The induction of apoptosis is, at least in part, due to a down-regulation of the expression of the antiapoptotic gene Bcl-XL. In addition to the thynic phenotype, RORgamma-/- mice lack lymph nodes, indicating that RORgamma is essential for lymph node organogenesis. Overexpression of RORgamma has been shown to inhibit T cell receptor-mediated apoptosis in T cell hybridomas and to repress the induction of Fas-ligand and interleukin 2. These studies demonstrate that RORs play critical roles in the regulation of a variety of physiological processes. Further characterization of the mechanisms of action of RORs will not only lead to the identification of ROR target genes and provide additional insight into their normal physiological functions, but will also determine their roles in disease.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Apoptosis
- Cloning, Molecular
- Gene Expression
- Hematopoiesis
- Humans
- Ligands
- Mice
- Mice, Knockout
- Molecular Sequence Data
- Nuclear Receptor Subfamily 1, Group F, Member 1
- Nuclear Receptor Subfamily 1, Group F, Member 2
- Nuclear Receptor Subfamily 1, Group F, Member 3
- Phenotype
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Receptors, Cytoplasmic and Nuclear/chemistry
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/physiology
- Receptors, Retinoic Acid
- Receptors, Thyroid Hormone
- Sequence Homology, Amino Acid
- Trans-Activators/chemistry
- Trans-Activators/genetics
- Trans-Activators/physiology
Collapse
Affiliation(s)
- A M Jetten
- Cell Biology Section, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, North Carolina 27709, USA.
| | | | | |
Collapse
|
50
|
Abstract
The estrogen receptor (ER) is a ligand-activated enhancer protein that is a member of the steroid/nuclear receptor superfamily. Two genes encode mammalian ER: ERalpha and ERbeta. ER binds to specific DNA sequences called estrogen response elements (EREs) with high affinity and transactivates gene expression in response to estradiol (E(2)). The purpose of this review is to summarize how natural and synthetic variations in the ERE sequence impact the affinity of ER-ERE binding and E(2)-induced transcriptional activity. Surprisingly, although the consensus ERE sequence was delineated in 1989, there are only seven natural EREs for which both ERalpha binding affinity and transcriptional activation have been examined. Even less information is available regarding how variations in ERE sequence impact ERbeta binding and transcriptional activity. Review of data from our own laboratory and those in the literature indicate that ERalpha binding affinity does not relate linearly with E(2)-induced transcriptional activation. We suggest that the reasons for this discord include cellular amounts of coactivators and adaptor proteins that play roles both in ER binding and transcriptional activation; phosphorylation of ER and other proteins involved in transcriptional activation; and sequence-specific and protein-induced alterations in chromatin architecture.
Collapse
Affiliation(s)
- C M Klinge
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|