101
|
Zhao C, Qiao Y, Jonsson P, Wang J, Xu L, Rouhi P, Sinha I, Cao Y, Williams C, Dahlman-Wright K. Genome-wide profiling of AP-1-regulated transcription provides insights into the invasiveness of triple-negative breast cancer. Cancer Res 2014; 74:3983-94. [PMID: 24830720 DOI: 10.1158/0008-5472.can-13-3396] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive clinical subtype accounting for up to 20% of all breast cancers, but its malignant determinants remain largely undefined. Here, we show that in TNBC the overexpression of Fra-1, a component of the transcription factor AP-1, offers prognostic potential. Fra-1 depletion or its heterodimeric partner c-Jun inhibits the proliferative and invasive phenotypes of TNBC cells in vitro. Similarly, RNAi-mediated attenuation of Fra-1 or c-Jun reduced cellular invasion in vivo in a zebrafish tumor xenograft model. Exploring the AP-1 cistrome and the AP-1-regulated transcriptome, we obtained insights into the transcriptional regulatory networks of AP-1 in TNBC cells. Among the direct targets identified for Fra-1/c-Jun involved in proliferation, adhesion, and cell-cell contact, we found that AP-1 repressed the expression of E-cadherin by transcriptional upregulation of ZEB2 to stimulate cell invasion. Overall, this work illuminates the pathways through which TNBC cells acquire invasive and proliferative properties.
Collapse
Affiliation(s)
- Chunyan Zhao
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge;
| | - Yichun Qiao
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge
| | - Philip Jonsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas; and
| | - Jian Wang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm
| | - Li Xu
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge
| | - Pegah Rouhi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm
| | - Indranil Sinha
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm; Department of Medicine and Health Sciences, Linköping University, Linköping; Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
| | - Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas; and
| | - Karin Dahlman-Wright
- Authors' Affiliations: Department of Biosciences and Nutrition, Novum, Karolinska Institute, Huddinge; Science for Life Laboratory, Karolinska Institute, Solna, Sweden;
| |
Collapse
|
102
|
Morrison G, Fu X, Shea M, Nanda S, Giuliano M, Wang T, Klinowska T, Osborne CK, Rimawi MF, Schiff R. Therapeutic potential of the dual EGFR/HER2 inhibitor AZD8931 in circumventing endocrine resistance. Breast Cancer Res Treat 2014; 144:263-72. [PMID: 24554387 PMCID: PMC4030601 DOI: 10.1007/s10549-014-2878-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 02/08/2014] [Indexed: 01/14/2023]
Abstract
Modest up-regulation of either HER-ligands or receptors has been implicated in acquired endocrine resistance. AZD8931, a dual tyrosine kinase inhibitor (TKI) of epithelial growth factor receptor (EGFR)/HER2, has been shown to more effectively block ligand-dependent HER signaling than the HER TKIs lapatinib or gefitinib. We therefore examined the effect of AZD8931 in ER-positive/HER2-negative breast cancer cells with acquired resistance to tamoxifen, where there is ligand up-regulation associated with HER pathway activation. RNA-seq ligand profiling and levels of HER receptors and signaling by western blotting were conducted in ER+ MCF7 and T47D parental cells and their Tam-resistant derivatives (TamRes). In vitro cell growth and apoptosis and HER ligand-stimulated signaling were measured in response to endocrine and HER TKIs. For studies in vivo, transplantable MCF7/TamRes xenografts were treated with tamoxifen or fulvestrant, either alone or in combination with AZD8931. AZD8931 only minimally enhanced endocrine sensitivity in MCF7 parental cells, but showed a greater effect in the T47D parental model. AZD8931 combined with either tamoxifen or fulvestrant inhibited cell growth more than lapatinib in T47D TamRes cells, and was also significantly, though modestly, more potent in MCF7 TamRes cells. In both TamRes models, AZD8931 significantly inhibited cell proliferation and induced apoptosis. Under ligand-stimulated conditions, AZD8931 more potently inhibited HER signaling than lapatinib or gefitinib. AZD8931 also significantly delayed the growth of MCF7 TamRes xenografts in the presence of tamoxifen or fulvestrant. The strongest inhibition was achieved with a fulvestrant and AZD8931 combination, though no tumor regression was observed. This study provides evidence that AZD8931 has greater inhibitory efficacy in tamoxifen-resistant settings than in an endocrine therapy naïve setting. The absence of tumor regression, however, suggests that additional escape pathways contribute to resistant growth and will need to be targeted to fully circumvent tamoxifen resistance.
Collapse
Affiliation(s)
- Gladys Morrison
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
- Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Xiaoyong Fu
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Marty Shea
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Sarmistha Nanda
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Mario Giuliano
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | - Tao Wang
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
| | | | - C. Kent Osborne
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
- Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Mothaffar F. Rimawi
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
- Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Rachel Schiff
- Lester and Sue Smith Breast Center and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030
- Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
103
|
Abstract
The nuclear receptor (NR) family comprises 48 transcription factors (TFs) with essential and diverse roles in development, metabolism and disease. Differently from other TFs, NRs engage with well-defined DNA-regulatory elements, mostly after ligand-induced structural changes. However, NR binding is not stochastic, and only a fraction of the cognate regulatory elements within the genome actively engage with NRs. In this review, we summarize recent advances in the understanding of the interactions between NRs and DNA. We discuss how chromatin accessibility and epigenetic modifications contribute to the recruitment and transactivation of NRs. Lastly, we present novel evidence of the interplay between non-coding RNA and NRs in the mediation of the assembly of the transcriptional machinery.
Collapse
Affiliation(s)
- Raffaella Maria Gadaleta
- Division of Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, W12 0NN, UK
| | | |
Collapse
|
104
|
Magnani L, Lupien M. Chromatin and epigenetic determinants of estrogen receptor alpha (ESR1) signaling. Mol Cell Endocrinol 2014; 382:633-641. [PMID: 23684889 DOI: 10.1016/j.mce.2013.04.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 04/16/2013] [Accepted: 04/29/2013] [Indexed: 12/30/2022]
Abstract
The oestrogen receptor alpha (ESR1) is a transcription factor that potentiates the response to diverse stimuli, including oestrogen and growth factors, in various tissue types. Its recruitment to the DNA is directly regulated by the chromatin landscape, inclusive of chromatin compaction and epigenetic modifications. In this review we discuss our current understanding of the interplay between ESR1 signaling and the chromatin landscape. We present how the chromatin landscape primes the lineage-specific response and contributes to stimuli-specific signaling. Finally, we discuss recent efforts to decipher the relationship between genetic and epigenetic as it relates to ESR1 signaling in breast cancer.
Collapse
Affiliation(s)
- Luca Magnani
- Ontario Cancer Institute, Princess Margaret Cancer Centre-University Health Network, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| | - Mathieu Lupien
- Ontario Cancer Institute, Princess Margaret Cancer Centre-University Health Network, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
105
|
Liu MH, Cheung E. Estrogen receptor-mediated long-range chromatin interactions and transcription in breast cancer. Mol Cell Endocrinol 2014; 382:624-632. [PMID: 24071518 DOI: 10.1016/j.mce.2013.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 09/17/2013] [Accepted: 09/17/2013] [Indexed: 12/15/2022]
Abstract
Estrogen induces the binding of ERα to thousands of locations in the breast cancer genome, preferring intergenic and distal regions rather than near the promoters of estrogen-regulated genes. With recent technological innovations in mapping and characterization of global chromatin organization, evidence now indicates ERα mediates long-range chromatin interactions to control gene transcription. The principles that govern how ERα communicates with their putative target genes via chromosomal interactions are also beginning to unravel. Herein, we summarize our current knowledge on the functional significance of chromatin looping in estrogen-mediated transcription. ERα collaborative factors and other players that contribute to define the genomic interactions in breast cancer cells will also be discussed. Defects in chromatin organization are emerging key players in diseases such as cancer, thus understanding how ERα-mediated chromatin looping affects genome organization will clarify the receptor's role in estrogen responsive pathways sensitive to defects in chromatin organization.
Collapse
Affiliation(s)
- Mei Hui Liu
- Food Science and Technology Programme, Department of Chemistry, National University of Singapore, Singapore 117543, Singapore; Cancer Biology and Pharmacology, Genome Institute of Singapore, A∗STAR (Agency for Science, Technology and Research), Singapore 138672, Singapore
| | - Edwin Cheung
- Cancer Biology and Pharmacology, Genome Institute of Singapore, A∗STAR (Agency for Science, Technology and Research), Singapore 138672, Singapore.
| |
Collapse
|
106
|
Patani N, Martin LA. Understanding response and resistance to oestrogen deprivation in ER-positive breast cancer. Mol Cell Endocrinol 2014; 382:683-694. [PMID: 24121024 DOI: 10.1016/j.mce.2013.09.038] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/30/2013] [Accepted: 09/30/2013] [Indexed: 01/01/2023]
Abstract
Oestrogens (E) and oestrogen receptor alpha (ERα) play fundamental roles in the development and progression of more than three-quarters of breast cancers (BC). The ability to influence the natural history of BC by hormonal manipulation is well established and endocrine therapies represent the cornerstone of systemic management for women with ERα-positive disease. Endocrine agents abrogate oestrogenic signalling through distinct and incompletely overlapping mechanisms, either impeding the transcriptional activity of ERα or diminishing E-synthesis. In post-menopausal women, E-production is chiefly attributable to the enzymatic conversion of androgens in extra-gonadal tissues by the cytochrome P-450 superfamily member aromatase. Greater understanding of steroid biosynthesis has underpinned rational drug design and pharmacological development of potent and specific aromatase inhibitors (AIs). Contemporary agents induce profound E-suppression in post-menopausal women and are first-line neo-adjuvant, adjuvant and metastatic therapies, with greater efficacy and tolerability than tamoxifen. The principal qualifier for endocrine treatment, including AIs, remains ERα expression. However, it is increasingly apparent that ERα expression is not synonymous with sensitivity to treatment and insufficient to account for the considerable heterogeneity of response. Better predictive biomarkers of de novo resistance are required to improve patient selection and identify those poor-responders who may benefit from alternative or additional systemic treatment from the outset. Among patients who do respond well initially, many relapse during their clinical course and there is also an unmet need for biomarkers of acquired resistance. The majority of women who relapse on AIs continue to express functional ERα which remains a legitimate target for second-line endocrine therapy. Understanding and overcoming acquired resistance to AIs requires a greater appreciation of ERα biology and the mechanisms though which E-dependence can be subverted. In this article, we review the impact of therapeutic E-deprivation on the natural history of ERα-positive breast cancer. Consideration is given to established and emerging biomarkers and/or determinants of response and resistance to E-deprivation. In vitro and in vivo evidence of the molecular mechanisms underpinning the transition from sensitivity to resistance are reviewed in the context of current models of ERα activity and their potential translational relevance.
Collapse
Affiliation(s)
- N Patani
- Academic Department of Biochemistry, Royal Marsden Foundation Trust, London, UK; Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JB, UK
| | - L-A Martin
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JB, UK.
| |
Collapse
|
107
|
Rangel LBA, Huang THM. Estrogen response in luminal breast cancer. Oncotarget 2013; 4:1548-9. [PMID: 24077736 PMCID: PMC3858541 DOI: 10.18632/oncotarget.1354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Leticia B A Rangel
- University of Texas Health Science Center at San Antonio, TX, USA and Universidade Federal do Espirito Santo, ES, Brazil/CNPq (Ciencias sem Fronteiras)
| | | |
Collapse
|
108
|
Fu X, Osborne CK, Schiff R. Biology and therapeutic potential of PI3K signaling in ER+/HER2-negative breast cancer. Breast 2013; 22 Suppl 2:S12-8. [PMID: 24011769 DOI: 10.1016/j.breast.2013.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PI3K is a central node mediating growth factor receptor signaling. With its downstream effectors such as AKT and mTOR, and its crosstalk with the RAS/RAF/MEK/MAPK pathway, it plays a vital role in cancer cell proliferation, metabolism, and survival. Recent breast cancer (BC) molecular portraits delineate PI3K as the most frequently altered pathway, with recurrent PIK3CA mutations mostly found in the luminal subtypes of BC. The transcriptomic and proteomic signatures of PI3K pathway activation associate with reduced estrogen receptor α (ER) levels and activity, and with the luminal B subtype of BC that has a relatively poor outcome. However, oncogenic transforming PIK3CA mutations have been shown to predict a better outcome in ER+/HER2-negative BC treated with endocrine therapy. In this review, we summarize the recent findings in the cause-and-effect of PI3K pathway aberration and endocrine sensitivity, especially the crosstalk with the ER pathway. Potential therapeutic approaches based on these findings are also discussed.
Collapse
Affiliation(s)
- Xiaoyong Fu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | |
Collapse
|
109
|
Everett LJ, Lazar MA. Cell-specific integration of nuclear receptor function at the genome. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2013; 5:615-29. [PMID: 23757177 PMCID: PMC3745817 DOI: 10.1002/wsbm.1231] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/01/2013] [Accepted: 05/02/2013] [Indexed: 11/08/2022]
Abstract
Nuclear receptors (NRs) encompass a family of regulatory proteins that directly couple small-molecule signaling to transcriptional regulation. Initial studies of specific NR targets led to a model in which NRs bind highly specific DNA motifs in proximal promoter regions and strongly induce gene transcription in response to ligand binding. More recently, genome-wide studies have added to the complexity of this classic model of NR function. In particular, binding of NRs at weaker or alternate motifs is common in the context of DNA assembled into chromatin, and ligand responsiveness varies at different NR target genes. Such findings have led to proposed modifications to the classic view of NR regulation, including the 'assisted loading' model in which NRs assist in opening chromatin rather than compete for binding sites, and context-specific models in which genomic and epigenomic features influence the NR function locally at each binding site. Further elucidation of these mechanisms will be particularly important for understanding cell-specific and ligand-specific functions of each NR. Emerging genomic technologies such as ChIP-seq and GRO-seq provide insights on a larger scale leading to deeper understanding of the complexities of transcriptional regulation by NRs.
Collapse
Affiliation(s)
- Logan J Everett
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
110
|
Zhang L, Cui J, Leonard M, Nephew K, Li Y, Zhang X. Silencing MED1 sensitizes breast cancer cells to pure anti-estrogen fulvestrant in vitro and in vivo. PLoS One 2013; 8:e70641. [PMID: 23936234 PMCID: PMC3728322 DOI: 10.1371/journal.pone.0070641] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/19/2013] [Indexed: 12/21/2022] Open
Abstract
Pure anti-estrogen fulvestrant has been shown to be a promising ER antagonist for locally advanced and metastatic breast cancer. Unfortunately, a significant proportion of patients developed resistance to this type of endocrine therapy but the molecular mechanisms governing cellular responsiveness to this agent remain poorly understood. Here, we’ve reported that knockdown of estrogen receptor coactivator MED1 sensitized fulvestrant resistance breast cancer cells to fulvestrant treatment. We found that MED1 knockdown further promoted cell cycle arrest induced by fulvestrant. Using an orthotopic xenograft mouse model, we found that knockdown of MED1 significantly reduced tumor growth in mice. Importantly, knockdown of MED1 further potentiated tumor growth inhibition by fulvestrant. Mechanistic studies indicated that combination of fulvestrant treatment and MED1 knockdown is able to cooperatively inhibit the expression of ER target genes. Chromatin immunoprecipitation experiments further supported a role for MED1 in regulating the recruitment of RNA polymerase II and transcriptional corepressor HDAC1 on endogenous ER target gene promoter in the presence of fulvestrant. These results demonstrate a role for MED1 in mediating resistance to the pure anti-estrogen fulvestrant both in vitro and in vivo.
Collapse
Affiliation(s)
- Lijiang Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Institute of Biochemistry, College of Life Science, Zhejiang University, Hangzhou City, China
- Center of Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou City, China
| | - Jiajun Cui
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Marissa Leonard
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Kenneth Nephew
- Department of Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana, United States of America
| | - Yongquan Li
- Institute of Biochemistry, College of Life Science, Zhejiang University, Hangzhou City, China
- * E-mail: (XZ); (YL)
| | - Xiaoting Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail: (XZ); (YL)
| |
Collapse
|
111
|
Eritja N, Mirantes C, Llobet D, Yeramian A, Bergadà L, Dosil MA, Domingo M, Matias-Guiu X, Dolcet X. Long-Term Estradiol Exposure Is a Direct Mitogen for Insulin/EGF-Primed Endometrial Cells and Drives PTEN Loss-Induced Hyperplasic Growth. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:277-87. [DOI: 10.1016/j.ajpath.2013.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/22/2013] [Accepted: 03/11/2013] [Indexed: 11/29/2022]
|
112
|
Ovaska K, Matarese F, Grote K, Charapitsa I, Cervera A, Liu C, Reid G, Seifert M, Stunnenberg HG, Hautaniemi S. Integrative analysis of deep sequencing data identifies estrogen receptor early response genes and links ATAD3B to poor survival in breast cancer. PLoS Comput Biol 2013; 9:e1003100. [PMID: 23818839 PMCID: PMC3688481 DOI: 10.1371/journal.pcbi.1003100] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 04/30/2013] [Indexed: 12/21/2022] Open
Abstract
Identification of responsive genes to an extra-cellular cue enables characterization of pathophysiologically crucial biological processes. Deep sequencing technologies provide a powerful means to identify responsive genes, which creates a need for computational methods able to analyze dynamic and multi-level deep sequencing data. To answer this need we introduce here a data-driven algorithm, SPINLONG, which is designed to search for genes that match the user-defined hypotheses or models. SPINLONG is applicable to various experimental setups measuring several molecular markers in parallel. To demonstrate the SPINLONG approach, we analyzed ChIP-seq data reporting PolII, estrogen receptor (), H3K4me3 and H2A.Z occupancy at five time points in the MCF-7 breast cancer cell line after estradiol stimulus. We obtained 777 early responsive genes and compared the biological functions of the genes having binding within 20 kb of the transcription start site (TSS) to genes without such binding site. Our results show that the non-genomic action of via the MAPK pathway, instead of direct binding, may be responsible for early cell responses to activation. Our results also indicate that the responsive genes triggered by the genomic pathway are transcribed faster than those without binding sites. The survival analysis of the 777 responsive genes with 150 primary breast cancer tumors and in two independent validation cohorts indicated the ATAD3B gene, which does not have binding site within 20 kb of its TSS, to be significantly associated with poor patient survival. Cellular processes in mammalian cells are tightly regulated to ensure that the cells function properly as a part of an organism. Dysregulation of some of these processes, such as apoptosis, cell proliferation and growth, can lead to cancer. One of the most important regulation mechanisms for cellular processes is via activation of membrane receptors by extra-cellular stimulus. Such cues trigger signal cascades that lead to altered expression of a number of genes in the cell nucleus; a key challenge in biomedicine is to identify which genes respond to a specific stimulus. These so called response genes can be investigated on a whole-genome scale with genomic sequencing, which is a technology that can quantify protein binding to DNA or gene activation. Analysis of such whole-genome data, however, is challenging due to billions of data points measured in the experiments. Here we introduce a novel computational method, SPINLONG, which is a widely applicable novel computational method that integrates multiple levels of deep sequencing data to produce experimentally testable hypotheses. We applied SPINLONG to breast cancer data and found early responsive genes for estrogen receptor and analyzed their regulation. These analyses resulted in a gene whose high activity is associated with decreased breast cancer patient survival.
Collapse
Affiliation(s)
- Kristian Ovaska
- Research Programs Unit, Genome-Scale Biology and Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Filomena Matarese
- Department of Molecular Biology, Faculty of Science, Nijmegen Center for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
| | | | | | - Alejandra Cervera
- Research Programs Unit, Genome-Scale Biology and Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Chengyu Liu
- Research Programs Unit, Genome-Scale Biology and Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - George Reid
- Institute of Molecular Biology, Mainz, Germany
| | | | - Hendrik G. Stunnenberg
- Department of Molecular Biology, Faculty of Science, Nijmegen Center for Molecular Life Sciences, Radboud University, Nijmegen, The Netherlands
| | - Sampsa Hautaniemi
- Research Programs Unit, Genome-Scale Biology and Institute of Biomedicine, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
113
|
Bhat-Nakshatri P, Song EK, Collins NR, Uversky VN, Dunker AK, O'Malley BW, Geistlinger TR, Carroll JS, Brown M, Nakshatri H. Interplay between estrogen receptor and AKT in estradiol-induced alternative splicing. BMC Med Genomics 2013; 6:21. [PMID: 23758675 PMCID: PMC3687557 DOI: 10.1186/1755-8794-6-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 06/03/2013] [Indexed: 02/07/2023] Open
Abstract
Background Alternative splicing is critical for generating complex proteomes in response to extracellular signals. Nuclear receptors including estrogen receptor alpha (ERα) and their ligands promote alternative splicing. The endogenous targets of ERα:estradiol (E2)-mediated alternative splicing and the influence of extracellular kinases that phosphorylate ERα on E2-induced splicing are unknown. Methods MCF-7 and its anti-estrogen derivatives were used for the majority of the assays. CD44 mini gene was used to measure the effect of E2 and AKT on alternative splicing. ExonHit array analysis was performed to identify E2 and AKT-regulated endogenous alternatively spliced apoptosis-related genes. Quantitative reverse transcription polymerase chain reaction was performed to verify alternative splicing. ERα binding to alternatively spliced genes was verified by chromatin immunoprecipitation assay. Bromodeoxyuridine incorporation-ELISA and Annexin V labeling assays were done to measure cell proliferation and apoptosis, respectively. Results We identified the targets of E2-induced alternative splicing and deconstructed some of the mechanisms surrounding E2-induced splicing by combining splice array with ERα cistrome and gene expression array. E2-induced alternatively spliced genes fall into at least two subgroups: coupled to E2-regulated transcription and ERα binding to the gene without an effect on rate of transcription. Further, AKT, which phosphorylates both ERα and splicing factors, influenced ERα:E2 dependent splicing in a gene-specific manner. Genes that are alternatively spliced include FAS/CD95, FGFR2, and AXIN-1. E2 increased the expression of FGFR2 C1 isoform but reduced C3 isoform at mRNA level. E2-induced alternative splicing of FAS and FGFR2 in MCF-7 cells correlated with resistance to FAS activation-induced apoptosis and response to keratinocyte growth factor (KGF), respectively. Resistance of MCF-7 breast cancer cells to the anti-estrogen tamoxifen was associated with ERα-dependent overexpression of FGFR2, whereas resistance to fulvestrant was associated with ERα-dependent isoform switching, which correlated with altered response to KGF. Conclusion E2 may partly alter cellular proteome through alternative splicing uncoupled to its effects on transcription initiation and aberration in E2-induced alternative splicing events may influence response to anti-estrogens.
Collapse
Affiliation(s)
- Poornima Bhat-Nakshatri
- Department of Surgery, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Magnani L, Brunelle M, Gévry N, Lupien M. Chromatin landscape and endocrine response in breast cancer. Epigenomics 2013; 4:675-83. [PMID: 23244312 DOI: 10.2217/epi.12.64] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Over two-thirds of breast cancers rely on estrogen receptor α (ERα) for their growth. Endocrine therapies antagonize estrogen-dependent ERα activation but resistance to these treatments occurs and is associated with poor prognosis. Crosstalk between alternative survival pathways and ERα are currently held as the primary cause of resistance. However, blocking these pathways does not cure endocrine therapy resistant breast cancer suggesting the existence of additional mechanisms. While cancer is commonly considered a genetic disease, the importance of epigenetic events in promoting tumor initiation and progression is increasingly recognized. Here, we consider how epigenetic modifications and alterations to the chromatin landscape contribute to endocrine therapy resistance by modulating ERα expression or altering its genomic activity.
Collapse
Affiliation(s)
- Luca Magnani
- Ontario Cancer Institute, Princess Margaret Hospital-University Health Network & the Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|
115
|
Morandi A, Martin LA, Gao Q, Pancholi S, Mackay A, Robertson D, Zvelebil M, Dowsett M, Plaza-Menacho I, Isacke CM. GDNF-RET signaling in ER-positive breast cancers is a key determinant of response and resistance to aromatase inhibitors. Cancer Res 2013; 73:3783-95. [PMID: 23650283 DOI: 10.1158/0008-5472.can-12-4265] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Most breast cancers at diagnosis are estrogen receptor-positive (ER(+)) and depend on estrogen for growth and survival. Blocking estrogen biosynthesis by aromatase inhibitors has therefore become a first-line endocrine therapy for postmenopausal women with ER(+) breast cancers. Despite providing substantial improvements in patient outcome, aromatase inhibitor resistance remains a major clinical challenge. The receptor tyrosine kinase, RET, and its coreceptor, GFRα1, are upregulated in a subset of ER(+) breast cancers, and the RET ligand, glial-derived neurotrophic factor (GDNF) is upregulated by inflammatory cytokines. Here, we report the findings of a multidisciplinary strategy to address the impact of GDNF-RET signaling in the response to aromatase inhibitor treatment. In breast cancer cells in two-dimensional and three-dimensional culture, GDNF-mediated RET signaling is enhanced in a model of aromatase inhibitor resistance. Furthermore, GDNF-RET signaling promoted the survival of aromatase inhibitor-resistant cells and elicited resistance in aromatase inhibitor-sensitive cells. Both these effects were selectively reverted by the RET kinase inhibitor, NVP-BBT594. Gene expression profiling in ER(+) cancers defined a proliferation-independent GDNF response signature that prognosed poor patient outcome and, more importantly, predicted poor response to aromatase inhibitor treatment with the development of resistance. We validated these findings by showing increased RET protein expression levels in an independent cohort of aromatase inhibitor-resistant patient specimens. Together, our results establish GDNF-RET signaling as a rational therapeutic target to combat or delay the onset of aromatase inhibitor resistance in breast cancer.
Collapse
Affiliation(s)
- Andrea Morandi
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, Department of Academic Biochemistry, The Royal Marsden Hospital; and London Research Institute, Cancer Research UK, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Zwart W, Koornstra R, Wesseling J, Rutgers E, Linn S, Carroll JS. A carrier-assisted ChIP-seq method for estrogen receptor-chromatin interactions from breast cancer core needle biopsy samples. BMC Genomics 2013; 14:232. [PMID: 23565824 PMCID: PMC3637562 DOI: 10.1186/1471-2164-14-232] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 03/22/2013] [Indexed: 01/16/2023] Open
Abstract
Background The Estrogen Receptor alpha (ERα) is the key transcriptional regulator in luminal breast cancer and is therefore the main target for adjuvant treatment of this subtype. Luminal gene signatures are dictated by the transcriptional capacities of ERα, which are a direct consequence of the receptors binding preference at specific sites on the chromatin. The identification of ERα binding signatures on a genome-wide level has greatly enhanced our understanding of Estrogen Receptor biology in cell lines and tumours, but the technique has its limitations with respect to its applicability in limited amounts of tumour tissue. Results Here, we present a refinement of the ChIP-seq procedures to enable transcription factor mapping on limited amounts of tissue culture cells as well as from a limited amount of tumor tissue derived from core needle biopsies. Our approach uses a carrier that can be removed prior to DNA amplification and sequencing. Conclusion We illustrate the applicability of this refined technology by mapping the ERα genome-wide chromatin binding landscape in core needle biopsy material from primary breast tumours. With this, our refined technology permits for a high-resolution transcription factor mapping even from clinical samples.
Collapse
Affiliation(s)
- Wilbert Zwart
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
117
|
Huang L, Tang Y, Xing D. Activation of nuclear estrogen receptors induced by low-power laser irradiation via PI3-K/Akt signaling cascade. J Cell Physiol 2013; 228:1045-59. [PMID: 23065720 DOI: 10.1002/jcp.24252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 09/26/2012] [Indexed: 12/23/2022]
Abstract
Low-power laser irradiation (LPLI) has been shown to exert promotive effects on cell survival and proliferation through activation of various signaling pathways. Estrogen receptors (ERs, ERα, and ERβ) are ligand-activated transcription factors, which regulate target gene expression, promote cell proliferation, and resist apoptosis. However, it is unclear whether LPLI could induce ligand-independent activation of ERs. In the present study, we investigated the subcellular pools, nuclear redistribution, and transcriptional activity of ERs under LPLI (1.2 J/cm(2), 633 nm) treatment using single-molecule fluorescence imaging and dual-luciferase reporter assay. We found that ERs were not only localized to nucleus, but also existed in mitochondria. Moreover, we found that LPLI induced nuclear redistribution and transcriptional activity of ERs in a ligand-independent manner. Our further investigation showed that PI3-K/Akt signaling cascade was involved in LPLI-induced activation of ERs. Wortmannin, a PI3-K inhibitor, or triciribine (API-2), a specific Akt inhibitor, potently suppressed the nuclear redistribution and transcriptional activity of ERs induced by LPLI, revealing that PI3-K/Akt signaling cascade was required for the activation of ERs induced by LPLI. Collectively, we demonstrated the first time that LPLI induced the ligand-independent nuclear redistribution and transcriptional activity of ERs, which were dependent on the activity of PI3-K/Akt. Our findings provide direct evidence for the molecular mechanisms of LPLI-induced transcription factor activation.
Collapse
Affiliation(s)
- Lei Huang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | | | | |
Collapse
|
118
|
Genome-wide reprogramming of the chromatin landscape underlies endocrine therapy resistance in breast cancer. Proc Natl Acad Sci U S A 2013; 110:E1490-9. [PMID: 23576735 DOI: 10.1073/pnas.1219992110] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The estrogen receptor (ER)α drives growth in two-thirds of all breast cancers. Several targeted therapies, collectively termed endocrine therapy, impinge on estrogen-induced ERα activation to block tumor growth. However, half of ERα-positive breast cancers are tolerant or acquire resistance to endocrine therapy. We demonstrate that genome-wide reprogramming of the chromatin landscape, defined by epigenomic maps for regulatory elements or transcriptional activation and chromatin openness, underlies resistance to endocrine therapy. This annotation reveals endocrine therapy-response specific regulatory networks where NOTCH pathway is overactivated in resistant breast cancer cells, whereas classical ERα signaling is epigenetically disengaged. Blocking NOTCH signaling abrogates growth of resistant breast cancer cells. Its activation state in primary breast tumors is a prognostic factor of resistance in endocrine treated patients. Overall, our work demonstrates that chromatin landscape reprogramming underlies changes in regulatory networks driving endocrine therapy resistance in breast cancer.
Collapse
|
119
|
Dunbier AK, Ghazoui Z, Anderson H, Salter J, Nerurkar A, Osin P, A'hern R, Miller WR, Smith IE, Dowsett M. Molecular profiling of aromatase inhibitor-treated postmenopausal breast tumors identifies immune-related correlates of resistance. Clin Cancer Res 2013; 19:2775-86. [PMID: 23493347 DOI: 10.1158/1078-0432.ccr-12-1000] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE Estrogen withdrawal by treatment with aromatase inhibitors is the most effective form of endocrine therapy for postmenopausal estrogen receptor-positive (ER+) breast cancer. However, response to therapy varies markedly and understanding of the precise molecular effects of aromatase inhibitors and causes of resistance is limited. We aimed to identify in clinical breast cancer those genes and pathways most associated with resistance to aromatase inhibitors by examining the global transcriptional effects of AI treatment. EXPERIMENTAL DESIGN Baseline and 2-week posttreatment biopsies were obtained from 112 postmenopausal women with ER+ breast cancer receiving neoadjuvant anastrozole. Gene expression data were obtained from 81 baseline and 2-week paired samples. Pathway analysis identified (i) the most prevalent changes in expression and (ii) the pretreatment genes/pathways most related to poor antiproliferative response. RESULTS A total of 1,327 genes were differentially expressed after 2-week treatment (false discovery rate < 0.01). Proliferation-associated genes and classical estrogen-dependent genes were strongly downregulated whereas collagens and chemokines were upregulated. Pretreatment expression of an inflammatory signature correlated with antiproliferative response to anastrozole and this observation was validated in an independent study. Higher expression of immune-related genes such as SLAMF8 and TNF as well as lymphocytic infiltration were associated with poorer response (P < 0.001) and validated in an independent cohort. CONCLUSIONS The molecular response to aromatase inhibitor treatment varies greatly between patients consistent with the variable clinical benefit from aromatase inhibitor treatment. Higher baseline expression of an inflammatory signature is associated with poor antiproliferative response and should be assessed further as a novel biomarker and potential target for aromatase inhibitor-treated patients.
Collapse
|
120
|
Biddie SC, John S. Minireview: Conversing with chromatin: the language of nuclear receptors. Mol Endocrinol 2013; 28:3-15. [PMID: 24196351 DOI: 10.1210/me.2013-1247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nuclear receptors are transcription factors that are activated by physiological stimuli to bind DNA in the context of chromatin and regulate complex biological pathways. Major advances in nuclear receptor biology have been aided by genome scale examinations of receptor interactions with chromatin. In this review, we summarize the roles of the chromatin landscape in regulating nuclear receptor function. Chromatin acts as a central integrator in the nuclear receptor-signaling axis, operating in distinct temporal modalities. Chromatin effects nuclear receptor action by specifying its genomic localization and interactions with regulatory elements. On receptor binding, changes in chromatin operate as an effector of receptor signaling to modulate transcriptional events. Chromatin is therefore an integral component of the pathways that guide nuclear receptor action in cell-type-specific and cell state-dependent manners.
Collapse
Affiliation(s)
- Simon C Biddie
- Addenbrooke's Hospital (S.C.B.), Cambridge University Hospitals National Health Service Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom; and National Institutes of Health (S.J.), National Cancer Institute, Laboratory for Genome Integrity, Bethesda, Maryland 20892
| | | |
Collapse
|
121
|
Hierarchical modularity in ERα transcriptional network is associated with distinct functions and implicates clinical outcomes. Sci Rep 2012; 2:875. [PMID: 23166858 PMCID: PMC3500769 DOI: 10.1038/srep00875] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 10/30/2012] [Indexed: 12/18/2022] Open
Abstract
Recent genome-wide profiling reveals highly complex regulation networks among ERα and its targets. We integrated estrogen (E2)-stimulated time-series ERα ChIP-seq and gene expression data to identify the ERα-centered transcription factor (TF) hubs and their target genes, and inferred the time-variant hierarchical network structures using a Bayesian multivariate modeling approach. With its recurrent motif patterns, we determined three embedded regulatory modules from the ERα core transcriptional network. The GO analyses revealed the distinct biological function associated with each of three embedded modules. The survival analysis showed the genes in each module were able to render a significant survival correlation in breast cancer patient cohorts. In summary, our Bayesian statistical modeling and modularity analysis not only reveals the dynamic properties of the ERα-centered regulatory network and associated distinct biological functions, but also provides a reliable and effective genomic analytical approach for the analysis of dynamic regulatory network for any given TF.
Collapse
|
122
|
Cui J, Germer K, Wu T, Wang J, Luo J, Wang SC, Wang Q, Zhang X. Cross-talk between HER2 and MED1 regulates tamoxifen resistance of human breast cancer cells. Cancer Res 2012; 72:5625-34. [PMID: 22964581 DOI: 10.1158/0008-5472.can-12-1305] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite the fact that most breast cancer patients have estrogen receptor (ER) α-positive tumors, up to 50% of the patients are or soon develop resistance to endocrine therapy. It is recognized that HER2 activation is one of the major mechanisms contributing to endocrine resistance. In this study, we report that the ER coactivator MED1 is a novel cross-talk point for the HER2 and ERα pathways. Tissue microarray analysis of human breast cancers revealed that MED1 expression positively correlates most strongly with HER2 status of the tumors. MED1 was highly phosphorylated, in a HER2-dependent manner, at the site known to be critical for its activation. Importantly, RNAi-mediated attenuation of MED1 sensitized HER2-overexpressing cells to tamoxifen treatment. MED1 and its phosphorylated form, but not the corepressors N-CoR and SMRT, were recruited to the ERα target gene promoter by tamoxifen in HER2-overexpressing cells. Significantly, MED1 attenuation or mutation of MED1 phosphorylation sites was sufficient to restore the promoter recruitment of N-CoR and SMRT. Notably, we found that MED1 is required for the expression of not only traditional E2-ERα target genes but also the newly described EGF-ERα target genes. Our results additionally indicated that MED1 is recruited to the HER2 gene and required for its expression. Taken together, these findings support a key role for MED1 in HER2-mediated tamoxifen resistance and suggest its potential usage as a therapeutic target to simultaneously block both ERα and HER2 pathways for the treatment of this type of endocrine resistant breast cancer.
Collapse
Affiliation(s)
- Jiajun Cui
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Meyer CA, Tang Q, Liu XS. Minireview: applications of next-generation sequencing on studies of nuclear receptor regulation and function. Mol Endocrinol 2012; 26:1651-9. [PMID: 22930692 DOI: 10.1210/me.2012-1150] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Next-generation sequencing technologies have expanded the experimental possibilities for studying the genome-wide regulation of transcription by nuclear receptors, their collaborating transcription factors, and coregulators. These technologies allow investigators to obtain abundance and DNA sequence information in a single experiment. In this review, we highlight proven and potential uses of next-generation sequencing in the study of gene regulation by nuclear receptors. We also provide suggestions on how to effectively leverage this technology in a collaborative environment.
Collapse
Affiliation(s)
- Clifford A Meyer
- Department of Biostatistics and Computational Biology, Harvard School of Public Health, Biostatistics and Computational Biology, 450 Brookline Street, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
124
|
Giuliano M, Schifp R, Osborne CK, Trivedi MV. Biological mechanisms and clinical implications of endocrine resistance in breast cancer. Breast 2012; 20 Suppl 3:S42-9. [PMID: 22015292 DOI: 10.1016/s0960-9776(11)70293-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endocrine therapy represents the first and most efficacious targeted treatment for women with estrogen receptor-positive (ER+) breast cancer. In the last four decades several hormonal agents have been successfully introduced in clinical practice as both palliative therapy for advanced disease and adjuvant treatment for prevention of tumor relapse. Nevertheless, the intrinsic and acquired resistance occurs in a significant proportion of patients, limiting the efficacy of endocrine treatments. Several molecular mechanisms have been proposed to be responsible for endocrine resistance. Loss of ER expression, altered activity of ER coregulators, deregulation of apoptosis and cell cycle signaling, and hyperactive receptor tyrosine kinase (RTK) and stress/cell kinase pathways can collectively orchestrate the development and sustenance of pharmacologic resistance to endocrine therapy. Preclinical and clinical evidence documents the plasticity in ER expression levels and signaling. As such, ER can either drive gene transcription and tumor progression directly or crosstalk with alternate RTK and cellular kinase signaling pathways, resulting in modulation of its own expression levels and transcriptional program. For this reason a therapeutic approach based on the combination of agents targeting both ER and RTK signaling represents a promising strategy to be tested. Among many RTKs, EGFR, HER2, and PI3K have been found to be viable targets for the combination therapy strategy, at least in the preclinical setting. However, early results from clinical trials testing combination strategies have been discordant, suggesting the need for better approaches to simultaneously inhibit multiple escape pathways and to select the patients who may benefit the most from these strategies.
Collapse
Affiliation(s)
- Mario Giuliano
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
125
|
PKA phosphorylation redirects ERα to promoters of a unique gene set to induce tamoxifen resistance. Oncogene 2012; 32:3543-51. [PMID: 22907427 DOI: 10.1038/onc.2012.361] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 06/18/2012] [Accepted: 06/30/2012] [Indexed: 01/12/2023]
Abstract
Protein kinase A (PKA)-induced estrogen receptor alpha (ERα) phosphorylation at serine residue 305 (ERαS305-P) can induce tamoxifen (TAM) resistance in breast cancer. How this phospho-modification affects ERα specificity and translates into TAM resistance is unclear. Here, we show that S305-P modification of ERα reprograms the receptor, redirecting it to new transcriptional start sites, thus modulating the transcriptome. By altering the chromatin-binding pattern, Ser305 phosphorylation of ERα translates into a 26-gene expression classifier that identifies breast cancer patients with a poor disease outcome after TAM treatment. MYC-target genes and networks were significantly enriched in this gene classifier that includes a number of selective targets for ERαS305-P. The enhanced expression of MYC increased cell proliferation in the presence of TAM. We demonstrate that activation of the PKA signaling pathway alters the transcriptome by redirecting ERα to new transcriptional start sites, resulting in altered transcription and TAM resistance.
Collapse
|
126
|
Garcia-Bassets I, Wang D. Cistrome plasticity and mechanisms of cistrome reprogramming. Cell Cycle 2012; 11:3199-210. [PMID: 22895178 DOI: 10.4161/cc.21281] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mammalian genomes contain thousands of cis-regulatory elements for each transcription factor (TF), but TFs only occupy a relatively small subset referred to as cistrome. Recent studies demonstrate that a TF cistrome might differ among different organisms, tissue types and individuals. In a cell, a TF cistrome might differ among different physiological states, pathological stages and between physiological and pathological conditions. It is, therefore, remarkable how highly plastic these binding profiles are, and how massively they can be reprogrammed in rapid response to intra/extracellular variations and during cell identity transitions and evolution. Biologically, cistrome reprogramming events tend to be followed by changes in transcriptional outputs, thus serving as transformative mechanisms to synchronically alter the biology of the cell. In this review, we discuss the molecular basis of cistrome plasticity and attempt to integrate the different mechanisms and biological conditions associated with cistrome reprogramming. Emerging data suggest that, when altered, these reprogramming events might be linked to tumor development and/or progression, which is a radical conceptual change in our mechanistic understanding of cancer and, potentially, other diseases.
Collapse
Affiliation(s)
- Ivan Garcia-Bassets
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | | |
Collapse
|
127
|
Bianco S, Gévry N. Endocrine resistance in breast cancer: from cellular signaling pathways to epigenetic mechanisms. Transcription 2012; 3:165-70. [PMID: 22771991 DOI: 10.4161/trns.20496] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although multiple cellular mechanisms have been proposed to explain endocrine resistance in breast cancer, the genomics events promoting the dysregulation of gene expression pattern are not clearly understood. Because chromatin plays a dynamic role in the estrogen receptor α (ERα) transcriptional program, we herein review signaling pathways implicated in endocrine resistance and try to merge them with recent epigenetic studies.
Collapse
Affiliation(s)
- Stéphanie Bianco
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | |
Collapse
|
128
|
Watters RJ, Benos PV, Oesterreich S. To bind or not to bind--FoxA1 determines estrogen receptor action in breast cancer progression. Breast Cancer Res 2012; 14:312. [PMID: 22713214 PMCID: PMC3446328 DOI: 10.1186/bcr3146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chromatin immunoprecipitation followed by massively parallel sequencing (ChIP-seq) is rapidly enabling the comprehensive characterization of genome-wide transcription factor-binding sites, thus defining the cistrome (cis-acting DNA targets of a trans-acting factor). Estrogen receptor (ER) ChIP-seq studies have been performed mainly in cell lines, but Ross-Innes and colleagues have now completed the first such study in clinical breast cancer samples. The study aimed at determining the dynamics of ER binding and differences between more and less aggressive primary breast tumors and metastases. The authors found that ER bound to DNA in both aggressive and drug-resistant tumors but to different sites and with different affinities. Given previous findings from cell lines, FoxA1 appears to play a critical role in this reprogramming of ER binding.
Collapse
Affiliation(s)
- Rebecca J Watters
- University of Pittsburgh Cancer Institute, Magee Women's Research Institute, 204 Craft Avenue, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
129
|
Integrative functional genomics identifies an enhancer looping to the SOX9 gene disrupted by the 17q24.3 prostate cancer risk locus. Genome Res 2012; 22:1437-46. [PMID: 22665440 PMCID: PMC3409257 DOI: 10.1101/gr.135665.111] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Genome-wide association studies (GWAS) are identifying genetic predisposition to various diseases. The 17q24.3 locus harbors the single nucleotide polymorphism (SNP) rs1859962 that is statistically associated with prostate cancer (PCa). It defines a 130-kb linkage disequilibrium (LD) block that lies in an ∼2-Mb gene desert area. The functional biology driving the risk associated with this LD block is unknown. Here, we integrate genome-wide chromatin landscape data sets, namely, epigenomes and chromatin openness from diverse cell types. This identifies a PCa-specific enhancer within the rs1859962 risk LD block that establishes a 1-Mb chromatin loop with the SOX9 gene. The rs8072254 and rs1859961 SNPs mapping to this enhancer impose allele-specific gene expression. The variant allele of rs8072254 facilitates androgen receptor (AR) binding driving increased enhancer activity. The variant allele of rs1859961 decreases FOXA1 binding while increasing AP-1 binding. The latter is key to imposing allele-specific gene expression. The rs8072254 variant in strong LD with the rs1859962 risk SNP can account for the risk associated with this locus, while rs1859961 is a rare variant less likely to contribute to the risk associated with this LD block. Together, our results demonstrate that multiple genetic variants mapping to a unique enhancer looping to the SOX9 oncogene can account for the risk associated with the PCa 17q24.3 locus. Allele-specific recruitment of the transcription factors androgen receptor (AR) and activating protein-1 (AP-1) account for the increased enhancer activity ascribed to this PCa-risk LD block. This further supports the notion that an integrative genomics approach can identify the functional biology disrupted by genetic risk variants.
Collapse
|
130
|
Gilfillan S, Fiorito E, Hurtado A. Functional genomic methods to study estrogen receptor activity. J Mammary Gland Biol Neoplasia 2012; 17:147-53. [PMID: 22588661 DOI: 10.1007/s10911-012-9254-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/03/2012] [Indexed: 02/06/2023] Open
Abstract
Estrogen Receptor (ER) is a nuclear receptor that mediates the actions of estrogen and tamoxifen. ER is expressed in a major fraction of human breast cancers. Recently, genomic maps for estrogen- and tamoxifen-ER have been published. Interestingly, estrogen and tamoxifen induce similar genomic interactions and both ligands have been shown to use co-operating factors. The interactions of these co-operating factors within ER regions have impact both on ER-DNA interactions and gene expression regulated by estrogen and tamoxifen. Moreover, the study of chromatin changes induced by these factors has also provided significant insight into our understanding of ER transcriptional regulation. This methods review describes some functional genomic methods to study the influence of both ER ligands and ER co-operating factors. The analysis of protein-DNA interactions and chromatin changes can be explored by using classical and novel methods such as Chromatin Immunoprecipitation (ChIP) or Formaldehyde-Assisted Isolation of Regulatory Elements (FAIRE). This review also explores the properties of each of these methods and the advantages of combining them with high throughput sequencing.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/metabolism
- Antineoplastic Agents, Hormonal/pharmacology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Chromatin Immunoprecipitation
- Cross-Linking Reagents/chemistry
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/metabolism
- Estrogens/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Genomics/methods
- Humans
- Ligands
- Mammary Glands, Human/drug effects
- Mammary Glands, Human/metabolism
- Neoplasm Proteins/chemistry
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Receptors, Estrogen/chemistry
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Regulatory Elements, Transcriptional/drug effects
- Response Elements/drug effects
- Selective Estrogen Receptor Modulators/metabolism
- Selective Estrogen Receptor Modulators/pharmacology
- Tamoxifen/metabolism
- Tamoxifen/pharmacology
Collapse
Affiliation(s)
- Siv Gilfillan
- Breast Cancer Research group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway, University of Oslo, Blindern, Oslo, Norway
| | | | | |
Collapse
|
131
|
Bugge A, Feng D, Everett LJ, Briggs ER, Mullican SE, Wang F, Jager J, Lazar MA. Rev-erbα and Rev-erbβ coordinately protect the circadian clock and normal metabolic function. Genes Dev 2012; 26:657-67. [PMID: 22474260 DOI: 10.1101/gad.186858.112] [Citation(s) in RCA: 397] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The nuclear receptor Rev-erbα regulates circadian rhythm and metabolism, but its effects are modest and it has been considered to be a secondary regulator of the cell-autonomous clock. Here we report that depletion of Rev-erbα together with closely related Rev-erbβ has dramatic effects on the cell-autonomous clock as well as hepatic lipid metabolism. Mouse embryonic fibroblasts were rendered arrhythmic by depletion of both Rev-erbs. In mouse livers, Rev-erbβ mRNA and protein levels oscillate with a diurnal pattern similar to that of Rev-erbα, and both Rev-erbs are recruited to a remarkably similar set of binding sites across the genome, enriched near metabolic genes. Depletion of both Rev-erbs in liver synergistically derepresses several metabolic genes as well as genes that control the positive limb of the molecular clock. Moreover, deficiency of both Rev-erbs causes marked hepatic steatosis, in contrast to relatively subtle changes upon loss of either subtype alone. These findings establish the two Rev-erbs as major regulators of both clock function and metabolism, displaying a level of subtype collaboration that is unusual among nuclear receptors but common among core clock proteins, protecting the organism from major perturbations in circadian and metabolic physiology.
Collapse
Affiliation(s)
- Anne Bugge
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Chand AL, Wijayakumara DD, Knower KC, Herridge KA, Howard TL, Lazarus KA, Clyne CD. The orphan nuclear receptor LRH-1 and ERα activate GREB1 expression to induce breast cancer cell proliferation. PLoS One 2012; 7:e31593. [PMID: 22359603 PMCID: PMC3281101 DOI: 10.1371/journal.pone.0031593] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 01/10/2012] [Indexed: 11/18/2022] Open
Abstract
Background Liver Receptor Homolog 1 (LRH-1, NR5A2) is an orphan nuclear receptor that is over-expressed in cancers in tissues such as the breast, colon and pancreas. LRH-1 plays important roles in embryonic development, steroidogenesis and cholesterol homeostasis. In tumor cells, LRH-1 induces proliferation and cell cycle progression. High LRH-1 expression is demonstrated in breast cancers, positively correlating with ERα status and aromatase activity. LRH-1 dependent cellular mechanisms in breast cancer epithelial cells are poorly defined. Hence in the present study we investigated the actions of LRH-1 in estrogen receptor α (ERα) positive breast cancer cells. Results The study aimed to investigate LRH-1 dependent mechanisms that promote breast cancer proliferation. We identified that LRH-1 regulated the expression of Growth Regulation by Estrogen in Breast Cancer 1 (GREB1) in MCF-7 and MDA-MB-231 cells. Over-expression of LRH-1 increased GREB1 mRNA levels while knockdown of LRH-1 reduced its expression. GREB1 is a well characterised ERα target gene, with three estrogen response elements (ERE) located on its promoter. Chromatin immunoprecipitation studies provided evidence of the co-localisation of LRH-1 and ERα at all three EREs. With electrophoretic mobility shift assays, we demonstrated direct binding of LRH-1 to EREs located on GREB1 and Trefoil Factor 1 (TFF1, pS2) promoters. LRH-1 and ERα co-operatively activated transcription of ERE luciferase reporter constructs suggesting an overlap in regulation of target genes in breast cancer cells. Over-expression of LRH-1 resulted in an increase in cell proliferation. This effect was more pronounced with estradiol treatment. In the presence of ICI 182,780, an ERα antagonist, LRH-1 still induced proliferation. Conclusions We conclude that in ER-positive breast cancer cells, LRH-1 promotes cell proliferation by enhancing ERα mediated transcription of target genes such as GREB-1. Collectively these findings indicate the importance of LRH-1 in the progression of hormone-dependent breast cancer and implicate LRH-1 as a potential avenue for drug development.
Collapse
Affiliation(s)
- Ashwini L Chand
- Prince Henry's Institute, Monash Medical Centre, Clayton, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
133
|
|
134
|
Gaughan EM, Come SE. Optimizing Endocrine Therapy for Metastatic Breast Cancer. CURRENT BREAST CANCER REPORTS 2012. [DOI: 10.1007/s12609-011-0063-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
135
|
Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 2012; 481:389-93. [PMID: 22217937 PMCID: PMC3272464 DOI: 10.1038/nature10730] [Citation(s) in RCA: 1421] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 11/23/2011] [Indexed: 12/22/2022]
Abstract
Oestrogen receptor-α (ER) is the defining and driving transcription factor in the majority of breast cancers and its target genes dictate cell growth and endocrine response, yet genomic understanding of ER function has been restricted to model systems. Here we map genome-wide ER-binding events, by chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq), in primary breast cancers from patients with different clinical outcomes and in distant ER-positive metastases. We find that drug-resistant cancers still recruit ER to the chromatin, but that ER binding is a dynamic process, with the acquisition of unique ER-binding regions in tumours from patients that are likely to relapse. The acquired ER regulatory regions associated with poor clinical outcome observed in primary tumours reveal gene signatures that predict clinical outcome in ER-positive disease exclusively. We find that the differential ER-binding programme observed in tumours from patients with poor outcome is not due to the selection of a rare subpopulation of cells, but is due to the FOXA1-mediated reprogramming of ER binding on a rapid timescale. The parallel redistribution of ER and FOXA1 binding events in drug-resistant cellular contexts is supported by histological co-expression of ER and FOXA1 in metastatic samples. By establishing transcription-factor mapping in primary tumour material, we show that there is plasticity in ER-binding capacity, with distinct combinations of cis-regulatory elements linked with the different clinical outcomes.
Collapse
|
136
|
Zaret KS, Carroll JS. Pioneer transcription factors: establishing competence for gene expression. Genes Dev 2011; 25:2227-41. [PMID: 22056668 DOI: 10.1101/gad.176826.111] [Citation(s) in RCA: 1156] [Impact Index Per Article: 82.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transcription factors are adaptor molecules that detect regulatory sequences in the DNA and target the assembly of protein complexes that control gene expression. Yet much of the DNA in the eukaryotic cell is in nucleosomes and thereby occluded by histones, and can be further occluded by higher-order chromatin structures and repressor complexes. Indeed, genome-wide location analyses have revealed that, for all transcription factors tested, the vast majority of potential DNA-binding sites are unoccupied, demonstrating the inaccessibility of most of the nuclear DNA. This raises the question of how target sites at silent genes become bound de novo by transcription factors, thereby initiating regulatory events in chromatin. Binding cooperativity can be sufficient for many kinds of factors to simultaneously engage a target site in chromatin and activate gene expression. However, in cases in which the binding of a series of factors is sequential in time and thus not initially cooperative, special "pioneer transcription factors" can be the first to engage target sites in chromatin. Such initial binding can passively enhance transcription by reducing the number of additional factors that are needed to bind the DNA, culminating in activation. In addition, pioneer factor binding can actively open up the local chromatin and directly make it competent for other factors to bind. Passive and active roles for the pioneer factor FoxA occur in embryonic development, steroid hormone induction, and human cancers. Herein we review the field and describe how pioneer factors may enable cellular reprogramming.
Collapse
Affiliation(s)
- Kenneth S Zaret
- Epigenetics Program, Institute for Regenerative Medicine, Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, USA.
| | | |
Collapse
|
137
|
Magnani L, Ballantyne EB, Zhang X, Lupien M. PBX1 genomic pioneer function drives ERα signaling underlying progression in breast cancer. PLoS Genet 2011; 7:e1002368. [PMID: 22125492 PMCID: PMC3219601 DOI: 10.1371/journal.pgen.1002368] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 09/18/2011] [Indexed: 01/19/2023] Open
Abstract
Altered transcriptional programs are a hallmark of diseases, yet how these are established is still ill-defined. PBX1 is a TALE homeodomain protein involved in the development of different types of cancers. The estrogen receptor alpha (ERα) is central to the development of two-thirds of all breast cancers. Here we demonstrate that PBX1 acts as a pioneer factor and is essential for the ERα-mediated transcriptional response driving aggressive tumors in breast cancer. Indeed, PBX1 expression correlates with ERα in primary breast tumors, and breast cancer cells depleted of PBX1 no longer proliferate following estrogen stimulation. Profiling PBX1 recruitment and chromatin accessibility across the genome of breast cancer cells through ChIP-seq and FAIRE-seq reveals that PBX1 is loaded and promotes chromatin openness at specific genomic locations through its capacity to read specific epigenetic signatures. Accordingly, PBX1 guides ERα recruitment to a specific subset of sites. Expression profiling studies demonstrate that PBX1 controls over 70% of the estrogen response. More importantly, the PBX1-dependent transcriptional program is associated with poor-outcome in breast cancer patients. Correspondingly, PBX1 expression alone can discriminate a priori the outcome in ERα-positive breast cancer patients. These features are markedly different from the previously characterized ERα-associated pioneer factor FoxA1. Indeed, PBX1 is the only pioneer factor identified to date that discriminates outcome such as metastasis in ERα-positive breast cancer patients. Together our results reveal that PBX1 is a novel pioneer factor defining aggressive ERα-positive breast tumors, as it guides ERα genomic activity to unique genomic regions promoting a transcriptional program favorable to breast cancer progression.
Collapse
Affiliation(s)
- Luca Magnani
- Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Institute of Quantitative Biomedical Sciences, Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Elizabeth B. Ballantyne
- Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Institute of Quantitative Biomedical Sciences, Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Xiaoyang Zhang
- Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Institute of Quantitative Biomedical Sciences, Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| | - Mathieu Lupien
- Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
- Institute of Quantitative Biomedical Sciences, Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire, United States of America
| |
Collapse
|
138
|
Simultaneous Inhibition of Estrogen Receptor and the HER2 Pathway in Breast Cancer: Effects of HER2 Abundance. Transl Oncol 2011; 4:293-300. [PMID: 21966546 DOI: 10.1593/tlo.11127] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 05/20/2011] [Accepted: 05/23/2011] [Indexed: 01/16/2023] Open
Abstract
The estrogen receptor (ER) pathway and the epidermal growth factor receptor (EGFR) pathway play pivotal roles in breast cancer progression. Targeted therapies able to intercept ER or signaling downstream to EGFR and its kin, HER2, are routinely used to treat distinct groups of breast cancer patients. However, patient responses are limited by resistance to endocrine therapy, which may be due to compensatory HER2/EGFR signaling. This raises the possibility that simultaneous interception of HER2 and ER may enhance therapeutic efficacy. To address the question, we treated breast cancer cells with both fulvestrant (ICI 182780), an ER antagonist with no agonist effects, and lapatinib, an orally available tyrosine kinase inhibitor specific to EGFR and HER2. Our results indicate that the combination of drugs is especially effective when applied to HER2-overexpressing, ER-positive cancer cells. Interestingly, fulvestrant activated the mitogen-activated protein kinase (MAPK) pathway of these cells, but complete inhibition of MAPK signaling was observed on cotreatment with lapatinib. Taken together, our observations reinforce the possibility that the effectiveness of combining anti-ER and anti-HER2/EGFR drugs may be especially effective on a relatively small subtype of HER2-overexpressing, ER-positive tumors of the breast.
Collapse
|
139
|
Métivier R. Are ‘omics of estrogen receptors defining potential targets for breast cancer treatment? Epigenomics 2011; 3:525-8. [DOI: 10.2217/epi.11.81] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Raphaël Métivier
- Equipe SP@RTE, Equipe labellisée ligue contre le Cancer, UMR CNRS 6026, Université de Rennes 1 Campus de Beaulieu, 35042 RENNES Cedex, France
| |
Collapse
|
140
|
Wang WM, Wu SY, Lee AY, Chiang CM. Binding site specificity and factor redundancy in activator protein-1-driven human papillomavirus chromatin-dependent transcription. J Biol Chem 2011; 286:40974-86. [PMID: 21937452 DOI: 10.1074/jbc.m111.290874] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Activator protein-1 (AP-1) regulates diverse gene responses triggered by environmental cues and virus-induced cellular stress. Although many signaling events leading to AP-1 activation have been described, the fundamental features underlying binding site selection and factor recruitment of dimeric AP-1 complexes to their target genes remain mostly uncharacterized. Using recombinant full-length human AP-1 dimers formed between c-Jun and Fos family members (c-Fos, FosB, Fra-1, Fra-2) for DNA binding and transcriptional analysis, we found that each of these AP-1 complex exhibits differential activity for distinct non-consensus AP-1 sites present in human papillomavirus (HPV), and each AP-1 complex is capable of activating transcription from in vitro-reconstituted HPV chromatin in a p300- and acetyl-CoA-dependent manner. Transcription from HPV chromatin requires AP-1-dependent and contact-driven recruitment of p300. Acetylation of dimeric AP-1 complexes by p300 enhances AP-1 binding to DNA. Using a human C-33A cervical cancer-derived cell line harboring the episomal HPV type 11 genome, we illustrate binding site selectivity recognized by c-Jun, JunB, JunD, and various Fos family members in a combinatorial and unique pattern, highlighting the diversity and importance of non-canonical binding site recognition by various AP-1 family proteins.
Collapse
Affiliation(s)
- Wei-Ming Wang
- Simmons Comprehensive Cancer Center, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
141
|
Miller TW, Balko JM, Fox EM, Ghazoui Z, Dunbier A, Anderson H, Dowsett M, Jiang A, Smith RA, Maira SM, Manning HC, González-Angulo AM, Mills GB, Higham C, Chanthaphaychith S, Kuba MG, Miller WR, Shyr Y, Arteaga CL. ERα-dependent E2F transcription can mediate resistance to estrogen deprivation in human breast cancer. Cancer Discov 2011; 1:338-51. [PMID: 22049316 PMCID: PMC3204388 DOI: 10.1158/2159-8290.cd-11-0101] [Citation(s) in RCA: 260] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Most estrogen receptor α (ER)-positive breast cancers initially respond to antiestrogens, but many eventually become estrogen-independent and recur. We identified an estrogen-independent role for ER and the CDK4/Rb/E2F transcriptional axis in the hormone-independent growth of breast cancer cells. ER downregulation with fulvestrant or small interfering RNA (siRNA) inhibited estrogen-independent growth. Chromatin immunoprecipitation identified ER genomic binding activity in estrogen-deprived cells and primary breast tumors treated with aromatase inhibitors. Gene expression profiling revealed an estrogen-independent, ER/E2F-directed transcriptional program. An E2F activation gene signature correlated with a lesser response to aromatase inhibitors in patients' tumors. siRNA screening showed that CDK4, an activator of E2F, is required for estrogen-independent cell growth. Long-term estrogen-deprived cells hyperactivate phosphatidylinositol 3-kinase (PI3K) independently of ER/E2F. Fulvestrant combined with the pan-PI3K inhibitor BKM120 induced regression of ER(+) xenografts. These data support further development of ER downregulators and CDK4 inhibitors, and their combination with PI3K inhibitors for treatment of antiestrogen-resistant breast cancers.
Collapse
Affiliation(s)
- Todd W. Miller
- Department of Cancer Biology, Vanderbilt University, Nashville, TN,Breast Cancer Research Program, Vanderbilt University, Nashville, TN
| | - Justin M. Balko
- Department of Medicine, Vanderbilt University, Nashville, TN
| | - Emily M. Fox
- Department of Medicine, Vanderbilt University, Nashville, TN
| | - Zara Ghazoui
- Breakthrough Breast Cancer Centre, Inst. of Cancer Research, Royal Marsden Hospital, London, UK
| | - Anita Dunbier
- Breakthrough Breast Cancer Centre, Inst. of Cancer Research, Royal Marsden Hospital, London, UK
| | - Helen Anderson
- Breakthrough Breast Cancer Centre, Inst. of Cancer Research, Royal Marsden Hospital, London, UK
| | - Mitch Dowsett
- Breakthrough Breast Cancer Centre, Inst. of Cancer Research, Royal Marsden Hospital, London, UK,Academic Department of Biochemistry, Royal Marsden Hospital, London, UK
| | - Aixiang Jiang
- Department of Biostatistics, Vanderbilt University, Nashville, TN
| | - R. Adam Smith
- Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN,Vanderbilt-Ingram Cancer Center; Institute of Imaging Sciences; Vanderbilt University, Nashville, TN
| | - Sauveur-Michel Maira
- Novartis Institute for Biomedical Research, Oncology Disease Area, CH-4002 Basel, Switzerland
| | - H. Charles Manning
- Breast Cancer Research Program, Vanderbilt University, Nashville, TN,Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN,Vanderbilt-Ingram Cancer Center; Institute of Imaging Sciences; Vanderbilt University, Nashville, TN
| | - Ana M. González-Angulo
- Department of Breast Medical Oncology, University of Texas, M.D. Anderson Cancer Center, Houston, TX,Department of Systems Biology, University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Gordon B. Mills
- Department of Systems Biology, University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | | | | | - Maria G. Kuba
- Department of Pathology Vanderbilt University, Nashville, TN
| | - William R. Miller
- Breast Research Group, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Yu Shyr
- Breast Cancer Research Program, Vanderbilt University, Nashville, TN,Department of Biostatistics, Vanderbilt University, Nashville, TN
| | - Carlos L. Arteaga
- Department of Cancer Biology, Vanderbilt University, Nashville, TN,Breast Cancer Research Program, Vanderbilt University, Nashville, TN,Department of Medicine, Vanderbilt University, Nashville, TN
| |
Collapse
|
142
|
George CL, Lightman SL, Biddie SC. Transcription factor interactions in genomic nuclear receptor function. Epigenomics 2011; 3:471-85. [DOI: 10.2217/epi.11.66] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transcription factors (TF) regulate gene expression acting as DNA sequence-specific binding factors, orchestrating cofactor recruitment and assembly of the transcriptional machinery. Nuclear receptors, a ligand-inducible TF class, regulate a large proportion of the genome, yet achieve highly cell-specific and context-dependent transcription, despite their widespread expression. High-throughput genome-wide profiling of TF binding reveals a startling proportion of colocalized cell- and context-specific TF-binding patterns, implying TF interactions play a critical role in transcription. These interactions depend on the chromatin architecture, that predominantly acts to predetermine accessibility of TF-binding sites at regulatory elements. Here, we summarize recent findings that highlight the importance of combinatorial TF interactions in determining diverse nuclear receptor-mediated transcriptional responses, emphasizing the significance of chromatin structure in directing TF and nuclear receptor recruitment. Interactions between TFs are likely to be a general mechanism of regulatory factors, contributing to transcriptional control in health and disease.
Collapse
Affiliation(s)
- Charlotte L George
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Faculty of Medicine & Dentistry, University of Bristol, Bristol, BS1 3NY, UK
| | - Stafford L Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Faculty of Medicine & Dentistry, University of Bristol, Bristol, BS1 3NY, UK
| | - Simon C Biddie
- Dorothy Hodgkin Building, Whitson Street, University of Bristol, Bristol, BS1 3NY, UK
| |
Collapse
|
143
|
Scott SM, Brown M, Come SE. Emerging data on the efficacy and safety of fulvestrant, a unique antiestrogen therapy for advanced breast cancer. Expert Opin Drug Saf 2011; 10:819-26. [PMID: 21699443 DOI: 10.1517/14740338.2011.595560] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Fulvestrant is an antiestrogen therapy with a unique mechanism of action. Unlike the selective estrogen receptor modulator tamoxifen, fulvestrant has no known estrogen agonist activity and is considered a pure antiestrogen. Its primary mechanism of action is thought to result from downregulation of the estrogen receptor (ER). Considerable data have demonstrated the efficacy of fulvestrant in postmenopausal women with ER-positive advanced breast cancer, both in the first-line setting and following disease progression on tamoxifen or aromatase inhibitors. Recent studies report improved benefit with alternative dosing strategies. At all administration schedules, fulvestrant has an excellent safety profile with no significant adverse effects. AREAS COVERED This article provides a review of the mechanism of action of fulvestrant and the preclinical and clinical data evaluating its use as a form of endocrine therapy. The reader will gain insight into the pharmacologic properties of the drug and its role in the treatment of advanced hormone receptor-positive breast cancer in postmenopausal women. EXPERT OPINION Based on data demonstrating the efficacy of fulvestrant, including prolonged clinical benefit in many patients, this well-tolerated antiestrogen is an important therapy for breast cancer. The optimal position of fulvestrant in the sequence of endocrine therapies for postmenopausal women and its role in combination regimens are not yet resolved.
Collapse
Affiliation(s)
- Sarah M Scott
- Beth Israel Deaconess Medical Center, Dana Farber/Harvard Cancer Center, 330 Brookline Ave, Boston, MA 02215, USA
| | | | | |
Collapse
|
144
|
More on FOX News: FOXA1 on the horizon of estrogen receptor function and endocrine response. Breast Cancer Res 2011; 13:307. [PMID: 21575280 PMCID: PMC3219191 DOI: 10.1186/bcr2849] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor α (ER) is a major driver of breast cancer and the target of endocrine therapy. Full disclosure of the cofactors regulating ER interactions with chromatin and its transcriptional regulatory activity is still elusive. Novel genome-wide profiling tools have mapped ER binding events in breast cancer cells and delineated cofactors important in ER activity. Among these, the Forkhead protein FOXA1 is emerging as a key factor dictating global chromatin structure and the transcriptional function of ER in breast and non-breast cancer cells. The significance of FOXA1 in the chromatin interactions and transcriptional regulation of both estrogen- and tamoxifen-bound ER, and in supporting tamoxifen-resistant cell growth, may impact current endocrine therapies.
Collapse
|