Oestrogen Receptor α AF-1 and AF-2 Domains Have Cell

Total Page:16

File Type:pdf, Size:1020Kb

Oestrogen Receptor α AF-1 and AF-2 Domains Have Cell ARTICLE DOI: 10.1038/s41467-018-07175-0 OPEN Oestrogen receptor α AF-1 and AF-2 domains have cell population-specific functions in the mammary epithelium Stéphanie Cagnet1, Dalya Ataca 1, George Sflomos1, Patrick Aouad1, Sonia Schuepbach-Mallepell2, Henry Hugues3, Andrée Krust4, Ayyakkannu Ayyanan1, Valentina Scabia1 & Cathrin Brisken 1 α α 1234567890():,; Oestrogen receptor (ER ) is a transcription factor with ligand-independent and ligand- dependent activation functions (AF)-1 and -2. Oestrogens control postnatal mammary gland development acting on a subset of mammary epithelial cells (MECs), termed sensor cells, which are ERα-positive by immunohistochemistry (IHC) and secrete paracrine factors, which stimulate ERα-negative responder cells. Here we show that deletion of AF-1 or AF-2 blocks pubertal ductal growth and subsequent development because both are required for expres- sion of essential paracrine mediators. Thirty percent of the luminal cells are ERα-negative by IHC but express Esr1 transcripts. This low level ERα expression through AF-2 is essential for cell expansion during puberty and growth-inhibitory during pregnancy. Cell-intrinsic ERα is not required for cell proliferation nor for secretory differentiation but controls transcript levels of cell motility and cell adhesion genes and a stem cell and epithelial mesenchymal transition (EMT) signature identifying ERα as a key regulator of mammary epithelial cell plasticity. 1 Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland. 2 Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland. 3 Centre Hospitalier Universitaire Vaudois, Department of Laboratory Medecine, University Hospital of Lausanne, CH-1011 Lausanne, Switzerland. 4 Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS UMR7104; INSERM U596; ULP, Collège de France) and Institut Clinique de la Souris, Illkirch, Strasbourg, France. These authors contributed equally: Stéphanie Cagnet, Dalya Ataca, George Sflomos. Correspondence and requests for materials should be addressed to C.B. (email: cathrin.brisken@epfl.ch) NATURE COMMUNICATIONS | (2018) 9:4723 | DOI: 10.1038/s41467-018-07175-0 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-07175-0 estrogens, 17β-estradiol (E2) and its metabolites, are extended beyond the sub-iliac lymph node to fill 61% of the fat Opivotal for the development and the physiology of the pad (Fig. 1b, c). In AF-10 and AF-20 littermates, no TEBs were breast and impinge on breast carcinogenesis. The oes- found and fat pad filling remained <3% (Fig. 1b, c). In adult, 8- to trogen receptor α (ERα) is expressed in 40% of the luminal cells 12-week-old WT females, fat pads were filled up to 80%, in their that make up the inner layer of the mammary epithelium sur- AF-10 and AF-20 littermates to 5.1% and 3% only, respectively rounded by basal/myoepithelial cells1. Oestrogens drive pubertal (Fig. 1c, Supplementary Figure 1b). In older WT females, which development in the mouse mammary gland and induce expres- have been exposed to repeated oestrous cycle related peaks of E2 sion of the progesterone receptor (PgR), activation of which and progesterone, side branching occurred (Supplementary Fig- drives cell proliferation during subsequent oestrous cycling and ure 1c, d; Fig. 1c) whereas the block of ductal growth persisted in pregnancy. Both hormones rely on paracrine factors to activate AF-20 females (Supplementary Figure 1b, c) as observed in stem cells and induce proliferation of other mammary epithelial ERα−/− females16. In older AF-10 females, few ducts occasionally cells (MECs)2. extended beyond the lymph node (Supplementary Figure 1c, d, The ERα belongs to the nuclear receptor family and is com- Fig. 1c). Mutant ducts were atrophic with decreased ductal dia- posed of six modular domains, namely, A to F3. Ligand- meters (Supplementary Figure 1c, d) but structurally intact with independent and ligand-dependent activation functions, AF-1 luminal and basal cell layers as revealed by histological analysis and AF-2 map to the A/B and E domains, respectively4,5. Ligand- (Supplementary Figure 1e). Immunohistochemistry (IHC) independent signalling results from phosphorylation of different revealed expression of ERα protein in AF-10 and AF-20 mammary serine residues in AF-1 by for instance MAPK6, GSK-37 or epithelia at levels comparable to WT controls (Fig. 1d), as cyclinA/cdk28. Upon activation, the receptor dimerises and reported for their uteri13,14. This excluded the possibility that the translocates to the nucleus where it interacts either directly with mutant ERα proteins were unstable and their expression in MECs the DNA via specific DNA sequences known as the oestrogen was reduced or lost. Thus, the phenotypes reflect the specific response elements, or indirectly via DNA-binding proteins like deletions of AF-1 or AF-2 domain and show that both are AP-19. Full ligand-dependent transcriptional activity relies on required for ERα function during ductal elongation. synergistic activities of AF-1 and AF-25. A small fraction of the ERα is found at the plasma membrane; it elicits rapid, non- Endocrine disturbances in AF-10 and AF-20 mice.AdultAF-10, genomic responses, which modulate multiple signalling pathways − − AF-20 and ERα / female mice were reported to have increased and create cross-talk between membrane and nuclear ERα10. serum levels of E2, luteinizing hormone and testosterone as mea- More than 70% of all breast cancers express the ERα and this is sured by radioimmunoassay17.Ourfinding that mammary gland exploited therapeutically. The most widely used agent, tamoxifen, development was already affected before puberty begged the ques- antagonises AF-211 and agonises AF-112, and is used in primary tion whether endocrine disturbances may occur earlier. As plasma and secondary breast cancer prevention. Most insights into the steroid hormone levels are very low prior to puberty, we used liquid molecular mechanisms underlying ERα signalling stem from chromatography tandem–mass spectrometry (LC–MS). We con- in vitro studies with ERα-positive (ERα+) breast cancer cell lines, firmed that both E2 and testosterone levels increased significantly in in particular MCF-7 cells which express very high levels of the all three mutants in adulthood. E2 was detected at levels of 12 pg/ml receptor and are exquisitely sensitive to E2. How ER signalling in plasma of WT females and increased almost 2-fold in AF-10 and occurs in vivo in normal and cancerous tissue is poorly under- − − threefold in AF-20 and ERα / females. Testosterone levels raised stood. To dissect the different aspects of ERα signalling in vivo, from on average 0.1 ng/ml in plasma of WT females to 0.7 ng/ml mice lacking specifically the AF-1 domain (AF-10)13 or the AF-2 − − plasma levels in AF-10, 1.5 ng/ml in AF-20 and 1.4 ng/ml in ERα / domain (ERαAF-20)14 were generated and compared to mice − − females (Fig. 2a). Plasma progesterone levels varied in WT females lacking the entire ERα (ERα / ); all three strains are viable but due to oestrous cycling not observed in the mutants in which have impaired reproductive functions, and distinct organ – ovarian cycles are not established. All three mutants had decreased defects13 15. The role of the AF-1 and the AF-2 domains in the − − 11-deoxycorticosterone levels while 17-OH-progesterone levels mammary epithelium was not analysed. Using ERα / mice, we − − were increased in ERα / females (Fig. 2a). have previously shown that ERα is required for ductal elongation Progesterone levels increased specifically in peripubertal AF-20 in the mammary epithelium16. females whereas androstenedione levels increased in peripubertal Here, we explore the role of AF-1 and AF-2 vs. intact ERα − − ERα / and pubertal AF-20 females (Fig. 2a). Testosterone was signalling in mammary gland development; we demonstrate dif- significantly increased in peripubertal AF-10 females. Hence, ferential roles that are dependent on cell type and/or ERα protein complex alterations of the endocrine milieu occurred already levels and uncover important functions of the ERα in apparently before and during puberty. ERα-luminal responder cells. An overall analysis of the average fold-changes in steroid levels in ERα mutants compared to WT and heterozygous littermates in Results relation to their biosynthetic pathways revealed comparable decrease of plasma levels of the progesterone derivatives, 11- Mammary gland development in ERαAF-10 and ERαAF-20 deoxycorticosterone and corticosterone, in all mutants in mice. To assess the impact of germ-line deletion of ERα ligand- adulthood (Fig. 2b). The levels of E2 and its precursors 17OH- dependent, AF-2, vs. ligand-independent, AF-1, genomic actions progesterone, androstenedione, and testosterone, were close to on mammary gland development, we analysed mammary glands − − WT levels in AF-10 mice but deviated similarly in ERα / and of AF-10 and AF-20 females and their respective WT littermates AF-20 females are in line with negative feedback loops of the E2 (Fig. 1a) at critical developmental stages using whole-mount biosynthetic pathway being regulated by ligand-dependent ER stereomicroscopy (Fig. 1b, Supplementary Figure 1a–d). Before activity. the onset of ovarian function, on postnatal day 21, all females had rudimentary ductal systems (Supplementary Figure 1a) with on average 4.7% fat pad filling in WT and <3% fat pad filling in
Recommended publications
  • Analysis of Estrogen Receptor Isoforms and Variants in Breast Cancer Cell Lines
    EXPERIMENTAL AND THERAPEUTIC MeDICINE 2: 537-544, 2011 Analysis of estrogen receptor isoforms and variants in breast cancer cell lines MAIE AL-BADER1, CHRISTOPHER FORD2, BUSHRA AL-AYADHY3 and ISSAM FRANCIS3 Departments of 1Physiology, 2Surgery, and 3Pathology, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait Received November 22, 2010; Accepted February 14, 2011 DOI: 10.3892/etm.2011.226 Abstract. In the present study, the expression of estrogen domain C, the DNA binding domain; domains D/E, bearing receptor (ER)α and ERβ isoforms in ER-positive (MCF7, both the activation function-2 (AF-2) and the ligand binding T-47D and ZR-75-1) and ER-negative (MDA-MB-231, SK-BR-3, domains; and finally, domain F, the C-terminal domain (6,7). MDA-MB-453 and HCC1954) breast cancer cell lines was The actions of estrogens are mediated by binding to ERs investigated. ERα mRNA was expressed in ER-positive and (ERα and/or ERβ). These receptors, which are co-expressed some ER-negative cell lines. ERα ∆3, ∆5 and ∆7 spliced in a number of tissues, form functional homodimers or variants were present in MCF7 and T-47D cells; ERα ∆5 heterodimers. When bound to estrogens as homodimers, the and ∆7 spliced variants were detected in ZR-75-1 cells. transcription of target genes is activated (8,9), while as heterodi- MDA-MB-231 and HCC1954 cells expressed ERα ∆5 and ∆7 mers, ERβ exhibits an inhibitory action on ERα-mediated gene spliced variants. The ERβ1 variant was expressed in all of the expression and, in many instances, opposes the actions of ERα cell lines and the ERβ2 variant in all of the ER-positive and (7,9).
    [Show full text]
  • Human and Mouse CD Marker Handbook Human and Mouse CD Marker Key Markers - Human Key Markers - Mouse
    Welcome to More Choice CD Marker Handbook For more information, please visit: Human bdbiosciences.com/eu/go/humancdmarkers Mouse bdbiosciences.com/eu/go/mousecdmarkers Human and Mouse CD Marker Handbook Human and Mouse CD Marker Key Markers - Human Key Markers - Mouse CD3 CD3 CD (cluster of differentiation) molecules are cell surface markers T Cell CD4 CD4 useful for the identification and characterization of leukocytes. The CD CD8 CD8 nomenclature was developed and is maintained through the HLDA (Human Leukocyte Differentiation Antigens) workshop started in 1982. CD45R/B220 CD19 CD19 The goal is to provide standardization of monoclonal antibodies to B Cell CD20 CD22 (B cell activation marker) human antigens across laboratories. To characterize or “workshop” the antibodies, multiple laboratories carry out blind analyses of antibodies. These results independently validate antibody specificity. CD11c CD11c Dendritic Cell CD123 CD123 While the CD nomenclature has been developed for use with human antigens, it is applied to corresponding mouse antigens as well as antigens from other species. However, the mouse and other species NK Cell CD56 CD335 (NKp46) antibodies are not tested by HLDA. Human CD markers were reviewed by the HLDA. New CD markers Stem Cell/ CD34 CD34 were established at the HLDA9 meeting held in Barcelona in 2010. For Precursor hematopoetic stem cell only hematopoetic stem cell only additional information and CD markers please visit www.hcdm.org. Macrophage/ CD14 CD11b/ Mac-1 Monocyte CD33 Ly-71 (F4/80) CD66b Granulocyte CD66b Gr-1/Ly6G Ly6C CD41 CD41 CD61 (Integrin b3) CD61 Platelet CD9 CD62 CD62P (activated platelets) CD235a CD235a Erythrocyte Ter-119 CD146 MECA-32 CD106 CD146 Endothelial Cell CD31 CD62E (activated endothelial cells) Epithelial Cell CD236 CD326 (EPCAM1) For Research Use Only.
    [Show full text]
  • Amplitude Modulation of Androgen Signaling by C-MYC
    Downloaded from genesdev.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Amplitude modulation of androgen signaling by c-MYC Min Ni,1,2 Yiwen Chen,3,4 Teng Fei,1,2 Dan Li,1,2 Elgene Lim,1,2 X. Shirley Liu,3,4,5 and Myles Brown1,2,5 1Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA; 2Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA; 3Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA; 4Harvard School of Public Health, Boston, Massachusetts 02215, USA Androgen-stimulated growth of the molecular apocrine breast cancer subtype is mediated by an androgen receptor (AR)-regulated transcriptional program. However, the molecular details of this AR-centered regulatory network and the roles of other transcription factors that cooperate with AR in the network remain elusive. Here we report a positive feed-forward loop that enhances breast cancer growth involving AR, AR coregulators, and downstream target genes. In the absence of an androgen signal, TCF7L2 interacts with FOXA1 at AR-binding sites and represses the basal expression of AR target genes, including MYC. Direct AR regulation of MYC cooperates with AR-mediated activation of HER2/HER3 signaling. HER2/HER3 signaling increases the transcriptional activity of MYC through phosphorylation of MAD1, leading to increased levels of MYC/MAX heterodimers. MYC in turn reinforces the transcriptional activation of androgen-responsive genes. These results reveal a novel regulatory network in molecular apocrine breast cancers regulated by androgen and AR in which MYC plays a central role as both a key target and a cooperating transcription factor to drive oncogenic growth.
    [Show full text]
  • An Animal Model with a Cardiomyocyte-Specific Deletion of Estrogen Receptor Alpha: Functional, Metabolic, and Differential Netwo
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2014 An animal model with a cardiomyocyte-specific deletion of estrogen receptor alpha: Functional, metabolic, and differential network analysis Sriram Devanathan Washington University School of Medicine in St. Louis Timothy Whitehead Washington University School of Medicine in St. Louis George G. Schweitzer Washington University School of Medicine in St. Louis Nicole Fettig Washington University School of Medicine in St. Louis Attila Kovacs Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Devanathan, Sriram; Whitehead, Timothy; Schweitzer, George G.; Fettig, Nicole; Kovacs, Attila; Korach, Kenneth S.; Finck, Brian N.; and Shoghi, Kooresh I., ,"An animal model with a cardiomyocyte-specific deletion of estrogen receptor alpha: Functional, metabolic, and differential network analysis." PLoS One.9,7. e101900. (2014). https://digitalcommons.wustl.edu/open_access_pubs/3326 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Sriram Devanathan, Timothy Whitehead, George G. Schweitzer, Nicole Fettig, Attila Kovacs, Kenneth S. Korach, Brian N. Finck, and Kooresh I. Shoghi This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/3326 An Animal Model with a Cardiomyocyte-Specific Deletion of Estrogen Receptor Alpha: Functional, Metabolic, and Differential Network Analysis Sriram Devanathan1, Timothy Whitehead1, George G. Schweitzer2, Nicole Fettig1, Attila Kovacs3, Kenneth S.
    [Show full text]
  • Semaphorin 4D Promotes Skeletal Metastasis in Breast Cancer
    UCLA UCLA Previously Published Works Title Semaphorin 4D Promotes Skeletal Metastasis in Breast Cancer. Permalink https://escholarship.org/uc/item/1vj106ww Journal PloS one, 11(2) ISSN 1932-6203 Authors Yang, Ying-Hua Buhamrah, Asma Schneider, Abraham et al. Publication Date 2016 DOI 10.1371/journal.pone.0150151 Peer reviewed eScholarship.org Powered by the California Digital Library University of California RESEARCH ARTICLE Semaphorin 4D Promotes Skeletal Metastasis in Breast Cancer Ying-Hua Yang1, Asma Buhamrah1, Abraham Schneider1,2, Yi-Ling Lin3, Hua Zhou1, Amr Bugshan1, John R. Basile1,2* 1 Department of Oncology and Diagnostic Sciences, University of Maryland Dental School, Baltimore, Maryland, United States of America, 2 Greenebaum Cancer Center, Baltimore, Maryland, United States of America, 3 Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America * [email protected] Abstract Bone density is controlled by interactions between osteoclasts, which resorb bone, and osteoblasts, which deposit it. The semaphorins and their receptors, the plexins, originally shown to function in the immune system and to provide chemotactic cues for axon guid- ance, are now known to play a role in this process as well. Emerging data have identified OPEN ACCESS Semaphorin 4D (Sema4D) as a product of osteoclasts acting through its receptor Plexin-B1 on osteoblasts to inhibit their function, tipping the balance of bone homeostasis in favor of Citation: Yang Y-H, Buhamrah A, Schneider A, Lin Y- resorption. Breast cancers and other epithelial malignancies overexpress Sema4D, so we L, Zhou H, Bugshan A, et al. (2016) Semaphorin 4D Promotes Skeletal Metastasis in Breast Cancer.
    [Show full text]
  • Estrogen Receptor a Binds to Peroxisome Proliferator ^ Activated
    Human Cancer Biology Estrogen Receptor A Binds to Peroxisome Proliferator ^ Activated Receptor Response Element and Negatively Interferes with Peroxisome Proliferator ^ Activated Receptor ; Signaling in Breast Cancer Cells Daniela Bonofiglio,1Sabrina Gabriele,1Saveria Aquila,1Stefania Catalano,1Mariaelena Gentile,2 Emilia Middea,1Francesca Giordano,2 and Sebastiano Ando' 2,3 Abstract Purpose: The molecular mechanisms involved in the repressive effects exerted by estrogen receptors (ER) on peroxisome proliferator ^ activated receptor (PPAR) g^ mediated transcriptional activity remain to be elucidated. The aim of the present study was to provide new insight into the crosstalk between ERa and PPARg pathways in breast cancer cells. Experimental Design: Using MCF7 and HeLa cells as model systems, we did transient trans- fections and electrophoretic mobility shift assay and chromatin immunoprecipitation studies to evaluate the ability of ERa to influence PPAR response element ^ mediated transcription. A pos- sible direct interaction between ERa and PPARg was ascertained by coimmunoprecipitation assay, whereas their modulatory role in the phosphatidylinositol 3-kinase (PI3K)/AKT pathway was evaluated by determining PI3K activity and AKT phosphorylation. As a biological counter- part, we investigated the growth response to the cognate ligands of both receptors in hormone-dependent MCF7 breast cancer cells. Results:Our data show for the first time that ERa binds to PPAR response element and represses its transactivation. Moreover, we have documented the physical and functional interactions of ERa and PPARg, which also involve the p85 regulatory subunit of PI3K. Interestingly, ERa and PPARg pathways have an opposite effect on the regulation of the PI3K/AKT transduction cascade, explaining, at least in part, the divergent response exerted by the cognate ligands 17 h-estradiol and BRL49653on MCF7 cell proliferation.
    [Show full text]
  • Agonists and Knockdown of Estrogen Receptor Β Differentially Affect
    Schüler-Toprak et al. BMC Cancer (2016) 16:951 DOI 10.1186/s12885-016-2973-y RESEARCH ARTICLE Open Access Agonists and knockdown of estrogen receptor β differentially affect invasion of triple-negative breast cancer cells in vitro Susanne Schüler-Toprak1*, Julia Häring1, Elisabeth C. Inwald1, Christoph Moehle2, Olaf Ortmann1 and Oliver Treeck1 Abstract Background: Estrogen receptor β (ERβ) is expressed in the majority of invasive breast cancer cases, irrespective of their subtype, including triple-negative breast cancer (TNBC). Thus, ERβ might be a potential target for therapy of this challenging cancer type. In this in vitro study, we examined the role of ERβ in invasion of two triple-negative breast cancer cell lines. Methods: MDA-MB-231 and HS578T breast cancer cells were treated with the specific ERβ agonists ERB-041, WAY200070, Liquiritigenin and 3β-Adiol. Knockdown of ERβ expression was performed by means of siRNA transfection. Effects on cellular invasion were assessed in vitro by means of a modified Boyden chamber assay. Transcriptome analyses were performed using Affymetrix Human Gene 1.0 ST microarrays. Pathway and gene network analyses were performed by means of Genomatix and Ingenuity Pathway Analysis software. Results: Invasiveness of MBA-MB-231 and HS578T breast cancer cells decreased after treatment with ERβ agonists ERB-041 and WAY200070. Agonists Liquiritigenin and 3β-Adiol only reduced invasion of MDA-MB-231 cells. Knockdown of ERβ expression increased invasiveness of MDA-MB-231 cells about 3-fold. Transcriptome and pathway analyses revealed that ERβ knockdown led to activation of TGFβ signalling and induced expression of a network of genes with functions in extracellular matrix, tumor cell invasion and vitamin D3 metabolism.
    [Show full text]
  • Retinoid X Receptor Suppresses a Metastasis-Promoting Transcriptional Program in Myeloid Cells Via a Ligand-Insensitive Mechanism
    Retinoid X receptor suppresses a metastasis-promoting transcriptional program in myeloid cells via a ligand-insensitive mechanism Mate Kissa, Zsolt Czimmerera,b, Gergely Nagyb, Pawel Bieniasz-Krzywiecc,d, Manuel Ehlingc,d, Attila Papa, Szilard Poliskaa,e, Pal Botof, Petros Tzerpose, Attila Horvatha, Zsuzsanna Kolostyaka, Bence Danielg, Istvan Szatmarif, Massimiliano Mazzonec,d, and Laszlo Nagya,b,g,1 aNuclear Receptor Research Laboratory, Department of Biochemistry and Molecular Biology, University of Debrecen, 4032 Debrecen, Hungary; bMTA-DE “Lendület” Immunogenomics Research Group, University of Debrecen, 4032 Debrecen, Hungary; cLaboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, 3000 Leuven, Belgium; dLaboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; eUD-GenoMed Ltd., 4032 Debrecen, Hungary; fStem Cell Differentiation Laboratory, Department of Biochemistry and Molecular Biology, University of Debrecen, 4032 Debrecen, Hungary; and gGenomic Control of Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL 32827 Edited by Bert W. O’Malley, Baylor College of Medicine, Houston, TX, and approved August 18, 2017 (received for review January 18, 2017) Retinoid X receptor (RXR) regulates several key functions in myeloid and identified a network of RXR-bound enhancers that control cells, including inflammatory responses, phagocytosis, chemokine angiogenic genes including Vegfa, Hbegf, Litaf,andHipk2 (9). secretion, and proangiogenic activity. Its importance, however, in Activation of these enhancers by RXR induced a proangiogenic tumor-associated myeloid cells is unknown. In this study, we transcriptional program and phenotype (9). Interestingly, we demonstrate that deletion of RXR in myeloid cells enhances lung found that half of the 5,200 RXR-occupied genomic sites were metastasis formation while not affecting primary tumor growth.
    [Show full text]
  • The Structure-Function Relationship of Angular Estrogens and Estrogen Receptor Alpha to Initiate Estrogen-Induced Apoptosis in Breast Cancer Cells S
    Supplemental material to this article can be found at: http://molpharm.aspetjournals.org/content/suppl/2020/05/03/mol.120.119776.DC1 1521-0111/98/1/24–37$35.00 https://doi.org/10.1124/mol.120.119776 MOLECULAR PHARMACOLOGY Mol Pharmacol 98:24–37, July 2020 Copyright ª 2020 The Author(s) This is an open access article distributed under the CC BY Attribution 4.0 International license. The Structure-Function Relationship of Angular Estrogens and Estrogen Receptor Alpha to Initiate Estrogen-Induced Apoptosis in Breast Cancer Cells s Philipp Y. Maximov, Balkees Abderrahman, Yousef M. Hawsawi, Yue Chen, Charles E. Foulds, Antrix Jain, Anna Malovannaya, Ping Fan, Ramona F. Curpan, Ross Han, Sean W. Fanning, Bradley M. Broom, Daniela M. Quintana Rincon, Jeffery A. Greenland, Geoffrey L. Greene, and V. Craig Jordan Downloaded from Departments of Breast Medical Oncology (P.Y.M., B.A., P.F., D.M.Q.R., J.A.G., V.C.J.) and Computational Biology and Bioinformatics (B.M.B.), University of Texas, MD Anderson Cancer Center, Houston, Texas; King Faisal Specialist Hospital and Research (Gen.Org.), Research Center, Jeddah, Kingdom of Saudi Arabia (Y.M.H.); The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois (R.H., S.W.F., G.L.G.); Center for Precision Environmental Health and Department of Molecular and Cellular Biology (C.E.F.), Mass Spectrometry Proteomics Core (A.J., A.M.), Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core (A.M.), and Dan L. Duncan molpharm.aspetjournals.org
    [Show full text]
  • The WT1 Wilms' Tumor Suppressor Gene Product Interacts with Estrogen Receptor-Α and Regulates IGF-I Receptor Gene Transcripti
    135 The WT1 Wilms’ tumor suppressor gene product interacts with estrogen receptor- and regulates IGF-I receptor gene transcription in breast cancer cells Naama Reizner, Sharon Maor, Rive Sarfstein, Shirley Abramovitch, Wade V Welshons1, Edward M Curran1, Adrian V Lee2 and Haim Werner Department of Clinical Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel 1Department of Veterinary Biomedical Sciences, University of Missouri, Columbia, Missouri 65211, USA 2Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA (Requests for offprints should be addressed to H Werner; Email: [email protected]) Abstract The IGF-I receptor (IGF-IR) has an important role in breast cancer development and progression. Previous studies have suggested that the IGF-IR gene is negatively regulated by a number of transcription factors with tumor suppressor activity, including the Wilms’ tumor protein WT1. The present study was aimed at evaluating the hypothesis that IGF-IR gene transcription in breast cancer cells is under inhibitory control by WT1 and, furthermore, that the mechanism of action of WT1 involves functional and physical interactions with estrogen receptor- (ER). Results of transient coexpression experiments showed that all four predominant isoforms of WT1 (including or lacking alternatively spliced exons 5 and 9) repressed IGF-IR promoter activity by 39–49%. To examine the potential interplay between WT1 and ER in control of IGF-IR gene transcription we employed ER-depleted C4 cells that were generated by clonal selection of ER-positive MCF-7 cells that were maintained in estrogen-free conditions. IGF-IR levels in C4 cells were |43% of the values in MCF-7 cells whereas WT1 levels in C4 cells were 4·25-fold higher than in MCF-7.
    [Show full text]
  • Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association Between BMI and Adult-Onset Non- Atopic
    Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association between BMI and Adult-Onset Non- Atopic Asthma Ayoung Jeong 1,2, Medea Imboden 1,2, Akram Ghantous 3, Alexei Novoloaca 3, Anne-Elie Carsin 4,5,6, Manolis Kogevinas 4,5,6, Christian Schindler 1,2, Gianfranco Lovison 7, Zdenko Herceg 3, Cyrille Cuenin 3, Roel Vermeulen 8, Deborah Jarvis 9, André F. S. Amaral 9, Florian Kronenberg 10, Paolo Vineis 11,12 and Nicole Probst-Hensch 1,2,* 1 Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; [email protected] (A.J.); [email protected] (M.I.); [email protected] (C.S.) 2 Department of Public Health, University of Basel, 4001 Basel, Switzerland 3 International Agency for Research on Cancer, 69372 Lyon, France; [email protected] (A.G.); [email protected] (A.N.); [email protected] (Z.H.); [email protected] (C.C.) 4 ISGlobal, Barcelona Institute for Global Health, 08003 Barcelona, Spain; [email protected] (A.-E.C.); [email protected] (M.K.) 5 Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain 6 CIBER Epidemiología y Salud Pública (CIBERESP), 08005 Barcelona, Spain 7 Department of Economics, Business and Statistics, University of Palermo, 90128 Palermo, Italy; [email protected] 8 Environmental Epidemiology Division, Utrecht University, Institute for Risk Assessment Sciences, 3584CM Utrecht, Netherlands; [email protected] 9 Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College, SW3 6LR London, UK; [email protected] (D.J.); [email protected] (A.F.S.A.) 10 Division of Genetic Epidemiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; [email protected] 11 MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, W2 1PG London, UK; [email protected] 12 Italian Institute for Genomic Medicine (IIGM), 10126 Turin, Italy * Correspondence: [email protected]; Tel.: +41-61-284-8378 Int.
    [Show full text]
  • A High-Throughput Genome-Integrated Assay Reveals Spatial Dependencies Governing Tcf7l2 Binding
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.16.993204; this version posted March 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. A high-throughput genome-integrated assay reveals spatial dependencies governing Tcf7l2 binding Tomasz Szczesnik1,2,3, Lendy Chu4, Joshua W. K. Ho1,2,5, Richard Sherwood4,6,7 March 16, 2020 1 Author affiliations 1. Victor Chang Cardiac Research Institute, NSW 2010 Darlinghurst, Australia. 2. St Vincent’s Clinical School, University of New South Wales, NSW 2010 Darlinghurst, Australia. 3. Department of Biosystems Science and Engineering, ETH-Zürich, Mattenstrasse 26, Basel, 4058, Switzerland. 4. Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, MA 02115 Boston, USA. 5. School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. 6. Hubrecht Institute, 3584 CT Utrecht, the Netherlands. 7. Corresponding author, [email protected] 2 Summary Predicting where transcription factors bind in the genome from their in-vitro DNA binding affinity is confounded by the large number of possible interactions with nearby transcription factors. To characterise the binding logic for the Wnt effector transcription factor Tcf7l2, we have developed a high-throughput screening platform in which thousands of 99-bp synthesised DNA sequences are inserted into a specific genomic locus through CRISPR/Cas9-based homology-directed repair, followed by measurement of Tcf7l2 binding by DamID. Using this platform at two genomic loci in mouse embryonic stem cells, we show that while the binding of Tcf7l2 closely follows the in-vitro motif binding strength and is influenced by local chromatin accessibility, it is also strongly affected by the surrounding 99-bp of sequence.
    [Show full text]