P53: Protection Against Tumor Growth Beyond Effects on Cell Cycle and Apoptosis Xuyi Wang1,2, Evan R

Total Page:16

File Type:pdf, Size:1020Kb

P53: Protection Against Tumor Growth Beyond Effects on Cell Cycle and Apoptosis Xuyi Wang1,2, Evan R Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0563 Cancer Review Research p53: Protection against Tumor Growth beyond Effects on Cell Cycle and Apoptosis Xuyi Wang1,2, Evan R. Simpson1,3, and Kristy A. Brown1,2 The tumor suppressor p53 has established functions in cancer. lication by Wang and colleagues demonstrates for the first time Specifically, it has been shown to cause cell-cycle arrest and that p53 is a key negative regulator of aromatase and, hence, apoptosis in response to DNA damage. It is also one of the most estrogen production in the breast tumor microenvironment. It commonly mutated or silenced genes in cancer and for this reason goes further by demonstrating that an important regulator of has been extensively studied. Recently, the role of p53 has been aromatase, the obesity-associated and tumor-derived factor pros- shown to go beyond its effects on cell cycle and apoptosis, with taglandin E2, inhibits p53 in the breast adipose stroma. This effects on metabolism emerging as a key contributor to cancer review presents these findings in the context of established and growth in situations where p53 is lost. Beyond this, the role of p53 emerging roles of p53 and discusses possible implications for the in the tumor microenvironment is poorly understood. The pub- treatment of breast cancer. Cancer Res; 75(23); 5001–7. Ó2015 AACR. Background induces apoptosis. The mitochondrial pathway is mainly reg- ulated by p53 effector Bcl-2 proteins such as Bax (4) and PUMA Traditional tumor suppressor functions of p53 (5). In tumors with wild-type p53, p53 responses can be The tumor suppressor p53, encoded by the TP53 gene, is the inhibited by downregulating p53 activity or its effectors' activ- most commonly silenced or mutated gene in cancer, with 50% to þ ity. For instance, in estrogen receptor–positive (ER )breast 55% of human cancers having experienced the loss of wild-type cancers, ER prevents the p53-mediated apoptotic response by p53 activity (reviewed in ref. 1). Under normal conditions, p53 directly interacting with p53 (6). levels are low and, in some cases, undetectable. However, stress Although most p53 mutations result in inactivation or dys- signals such as DNA damage, oncogene activation, and hypoxia function of p53, some mutations in p53 lead to the selective loss stabilize p53 protein and induce increased cellular p53 levels by of apoptotic functions while retaining the ability to induce cell- posttranslational modifications such as phosphorylation and cycle arrest. Certain p53 mutations lead to p53 becoming onco- acetylation. Activated p53 causes a variety of responses including genic through gain-of-function mechanisms (7). Cell-cycle arrest cell-cycle arrest or apoptosis, thereby providing a critical barrier driven by p53 requires the transcription of p21, which is a cyclin- against tumor development (reviewed in refs. 1, 2). As a tran- dependent kinase inhibitor, or other p53 target genes such as scription factor, activated p53 binds to a number of genes that 14–3–3s and GADD45 (8). In general, DNA damage or stress will contain p53-binding sites within their regulatory regions. Bioin- increase levels of p53 protein, which in turn induces p21 tran- formatic studies have found more than 4,000 putative p53- scription and leads to cell-cycle arrest at G , allowing cells to binding sites in the existing human genome. Somatic mutations 1 survive until the damage has been repaired or the stress removed in TP53 are common in cancers and are associated with poor (9, 10). The G arrest is primarily regulated by p21, whereas G prognosis and low response to chemotherapy (3). 1 2 arrest is stimulated by GADD45, p21, and 14–3–3s (11). The role Numerous p53 target genes have been identified as down- of p53 to suppress tumor growth and promote apoptosis via these stream effectors of p53 with changesincellfunctionbeing pathways is well-characterized. However, novel roles for p53 are dependent on the regulation of several genes (2). For example, emerging and these are proving important contributors to the p53 mediates cell apoptosis by activating mitochondrial and tumor suppressor functions of p53. death receptor–induced apoptotic pathways, both pathways resulting in the induction of caspase signaling, which then p53 as a metabolic checkpoint Emerging evidence suggests that p53 is also involved in the regulation of metabolism and cell homeostasis without causing 1 Metabolism and Cancer Laboratory, Centres for Cancer Research and cell-cycle arrest or apoptosis (12). For example, nutrient deficien- Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia. 2Department of Physiology, Monash Uni- cy leads to the activation of p53 through direct phosphorylation at versity, Clayton, Victoria, Australia. 3Department of Biochemistry and Ser15 by AMP-activated protein kinase (AMPK), a key regulator of Molecular Biology, Monash University, Clayton, Victoria, Australia. cell metabolism (13). p53 also regulates metabolism and cell Corresponding Author: Kristy A. Brown, Metabolism and Cancer Laboratory, homeostasis in normal cells and tissues. Lipin1 is a recently Hudson Institute of Medical Research, 27–31 Wright Street, Clayton, Victoria identified p53 target gene that regulates the expression of genes 3168, Australia. Phone: 61-3-95944333; Fax: 61-3-95946125; E-mail: involved in fatty acid oxidation through PPARa (14). Under [email protected] nutrient/glucose deprivation conditions, p53 is upregulated by doi: 10.1158/0008-5472.CAN-15-0563 AMPK and stimulates Lipin1 and malonyl-CoA decarboxylase Ó2015 American Association for Cancer Research. expression, leading to an increase in fatty acid oxidation (14, 15). www.aacrjournals.org 5001 Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0563 Wang et al. This allows cells to use fatty acids as an alternative energy source. presence of FSK/PMA (30). This, in turn, drives the expression Reciprocally, p53 also promotes the expression of AMPK, leading of aromatase. We have also recently demonstrated that HIF1a to the negative regulation of mTOR (16). The PI3K/Akt/mTOR with known roles in promoting tumor growth and emerging pathway can suppress apoptosis and stimulate proinflammatory roles in regulating metabolism is stimulated by PGE2 in ASCs gene expression, which in turn promotes cancer growth and and stimulates the expression of aromatase. Unpublished data progression (17). Negative regulation of mTOR is also observed from Sasano and colleagues showed enhanced aromatase in autophagy, which can be induced by starvation and metabolic expression and staining in tightly packed undifferentiated ASCs stresses. around tumor cells in patients with breast cancer. This process Oncogenic transformation is often associated with enhanced of undifferentiation and increased stromal cell proliferation is aerobic glycolysis and reduced oxidative phosphorylation, which known as desmoplasia, which leads to the formation of a dense is known as the Warburg effect. Aerobic glycolysis allows cancer fibroblast layer surrounding malignant epithelial cells; it is cells to generate ATP as well as stimulate anaplerotic metabolism essential for structural and biochemical support of tumor of intermediates such as a-ketoglutarate to support cancer cell growth (31, 32). proliferation and survival (18). Recently, p53 has been shown to inhibit the Warburg effect by reducing glycolysis and enhancing Li–Fraumeni syndrome oxidative phosphorylation via upregulation of genes including Li–Fraumeni syndrome (LFS) is a rare autosomal dominant TIGAR and SCO2 (synthesis of cytochrome oxidase 2), as well as hereditary syndrome with the majority of affected individuals inhibiting the expression of glucose transporters GLUT1, GLUT3, carrying germ line mutations in the TP53 gene (33). LFS is and GLUT4 (19, 20). SCO2 increases mitochondrial respiration characterized by a high susceptibility of developing a number of and TIGAR inhibits glycolysis and promotes NADPH production malignancies, predominantly in childhood and early adult life. and glutathione recycling, whereas repression of glucose trans- Half of individuals with LFS develop at least one LFS-associated porters blocks the uptake of glucose (19). cancer by age 30 and 15% to 35% of cancer survivors with LFS will In summary, a number of genes are regulated by p53 to develop multiple primary tumors over their lifetimes (34). It has maintain metabolism and energy homeostasis in cells/tissues also been proposed that patients with LFS have deregulated under normal and stressed physiologic conditions. Changes in metabolism, including increased oxidative stress and hypoxia, to tumor cell metabolism are now recognized as a hallmark of cancer provide a microenvironment conducive to tumor formation, (21) and p53 is integral to this process. which can be one complication of dysfunctional p53 for these patients (35). Breast cancer is the most common type of cancer in Dysregulated metabolism as a driver of estrogen production in þ women with LFS and the majority of tumors are ER (36). the breast adipose Moll and colleagues previously found that wild-type p53 can Adipose tissue is responsible for energy storage and acts as an accumulate in the cytoplasm of tumor cells in inflammatory endocrine organ that regulates metabolism via endocrine, breast cancers, leading to functional inactivation
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]
  • Progesterone Receptor Coregulators As Factors Supporting the Function of the Corpus Luteum in Cows
    G C A T T A C G G C A T genes Article Progesterone Receptor Coregulators as Factors Supporting the Function of the Corpus Luteum in Cows Robert Rekawiecki * , Karolina Dobrzyn, Jan Kotwica and Magdalena K. Kowalik Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10–747 Olsztyn, Poland; [email protected] (K.D.); [email protected] (J.K.); [email protected] (M.K.K.) * Correspondence: [email protected]; Tel.: +48-89-539-31-17 Received: 5 July 2020; Accepted: 7 August 2020; Published: 12 August 2020 Abstract: Progesterone receptor (PGR) for its action required connection of the coregulatory proteins, including coactivators and corepressors. The former group exhibits a histone acetyltransferase (HAT) activity, while the latter cooperates with histone deacetylase (HDAC). Regulations of the coregulators mRNA and protein and HAT and HDAC activity can have an indirect effect on the PGR function and thus progesterone (P4) action on target cells. The highest mRNA expression levels for the coactivators—histone acetyltransferase p300 (P300), cAMP response element-binding protein (CREB), and steroid receptor coactivator-1 (SRC-1)—and nuclear receptor corepressor-2 (NCOR-2) were found in the corpus luteum (CL) on days 6 to 16 of the estrous cycle. The CREB protein level was higher on days 2–10, whereas SRC-1 and NCOR-2 were higher on days 2–5. The activity of HAT and HDAC was higher on days 6–10 of the estrous cycle. All of the coregulators were localized in the nuclei of small and large luteal cells.
    [Show full text]
  • Identification of a Different-Type Homeobox Gene, Barhi, Possibly Causing Bar (B) and Om(Ld) Mutations in Drosophila
    Proc. Nati. Acad. Sci. USA Vol. 88, pp. 4343-4347, May 1991 Genetics Identification of a different-type homeobox gene, BarHI, possibly causing Bar (B) and Om(lD) mutations in Drosophila (compound eye/homeodomain/morphogenesis/malformation/M-repeat) TETSUYA KOJIMA, SATOSHI ISHIMARU, SHIN-ICHI HIGASHUIMA, Eui TAKAYAMA, HIROSHI AKIMARU, MASAKI SONE, YASUFUMI EMORI, AND KAORU SAIGO* Department of Biophysics and Biochemistry, Faculty of Science, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113, Japan Communicated by Melvin M. Green, January 25, 1991 ABSTRACT The Bar mutation B of Drosophila melano- the initial periodicity. A class of mutations including Bar (B) gaster and optic morphology mutation Om(JD) of Drosophila may also be intimately related to early events (9). For ananassae result in suppression of ommatidium differentiation clarification of this point, examination was first made of at the anterior portion of the eye. Examination was made to possible determinant genes for the Bar mutation B of Dro- determine the genes responsible for these mutations. Both loci sophila melanogaster (10) and optic morphology mutation were found to share in common a different type of homeobox Om(JD) of Drosophila ananassae (11), in both of which gene, which we call "BarH1." Polypeptides encoded by D. ommatidium differentiation is suppressed in the anterior melanogaster and D. ananassae BarHl genes consist of543 and portion ofthe eye. Both locit were found to share in common 604 amino acids, respectively, with homeodomains identical in a different type of homeobox gene, called BarH1, which sequence except for one amino acid substitution. A unique encodes a polypeptide of Mr - 60,000.
    [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]
  • Etiology of Hormone Receptor–Defined Breast Cancer: a Systematic Review of the Literature
    1558 Cancer Epidemiology, Biomarkers & Prevention Review Etiology of Hormone Receptor–Defined Breast Cancer: A Systematic Review of the Literature Michelle D. Althuis, Jennifer H. Fergenbaum, Montserrat Garcia-Closas, Louise A. Brinton, M. Patricia Madigan, and Mark E. Sherman Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland Abstract Breast cancers classified by estrogen receptor (ER) and/ negative tumors. Postmenopausal obesity was also or progesterone receptor (PR) expression have different more consistently associated with increased risk of clinical, pathologic, and molecular features. We exam- hormone receptor–positive than hormone receptor– ined existing evidence from the epidemiologic litera- negative tumors, possibly reflecting increased estrogen ture as to whether breast cancers stratified by hormone synthesis in adipose stores and greater bioavailability. receptor status are also etiologically distinct diseases. Published data are insufficient to suggest that exoge- Despite limited statistical power and nonstandardized nous estrogen use (oral contraceptives or hormone re- receptor assays, in aggregate, the critically evaluated placement therapy) increase risk of hormone-sensitive studies (n = 31) suggest that the etiology of hormone tumors. Risks associated with breast-feeding, alcohol receptor–defined breast cancers may be heterogeneous. consumption, cigarette smoking, family history of Reproduction-related exposures tended to be associat-
    [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]
  • Estrogen Receptor-Α Interaction with the CREB Binding Protein
    307 Estrogen receptor- interaction with the CREB binding protein coactivator is regulated by the cellular environment B M Jaber, R Mukopadhyay and C L Smith Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA (Requests for offprints should be addressed to C L Smith; Email: [email protected]) Abstract The p160 coactivators, steroid receptor coactivator-1 (SRC-1), transcriptional intermediary factor-2 (TIF2) and receptor-associated coactivator-3 (RAC3), as well as the coactivator/integrator CBP, mediate estrogen receptor- (ER)-dependent gene expression. Although these coactivators are widely expressed, ER transcriptional activity is cell-type dependent. In this study, we investigated ER interaction with p160 coactivators and CBP in HeLa and HepG2 cell lines. Basal and estradiol (E2)-dependent interactions between the ER ligand-binding domain (LBD) and SRC-1, TIF2 or RAC3 were observed in HeLa and HepG2 cells. The extents of hormone-dependent interactions were similar and interactions between each of the p160 coactivators and the ER LBD were not enhanced by 4-hydroxytamoxifen in either cell type. In contrast to the situation for p160 coactivators, E2-dependent interaction of the ER LBD with CBP or p300 was detected in HeLa but not HepG2 cells by mammalian two-hybrid and coimmunoprecipitation assays, indicating that the cellular environment modulates ER-CBP/p300 interaction. Furthermore, interactions between CBP and p160 coactivators are much more robust in HeLa than HepG2 cells suggesting that poor CBP-p160 interactions are insufficient to support ER–CBP–p160 ternary complexes important for nuclear receptor–CBP interactions. Alterations in p160 coactivators or CBP expression between these two cell types did not account for differences in ER–p160–CBP interactions.
    [Show full text]
  • Understanding Nuclear Receptor Form and Function Using Structural Biology
    F RASTINEJAD and others Understanding NR form 51:3 T1–T21 Thematic Review and function Understanding nuclear receptor form and function using structural biology Correspondence Fraydoon Rastinejad, Pengxiang Huang, Vikas Chandra and Sepideh Khorasanizadeh should be addressed to F Rastinejad Metabolic Signaling and Disease Program, Sanford-Burnham Medical Research Institute, Orlando, Email Florida 32827, USA frastinejad@ sanfordburnham.org Abstract Nuclear receptors (NRs) are a major transcription factor family whose members selectively Key Words bind small-molecule lipophilic ligands and transduce those signals into specific changes in " nuclear receptors gene programs. For over two decades, structural biology efforts were focused exclusively on " steroid hormones the individual ligand-binding domains (LBDs) or DNA-binding domains of NRs. These " transcription factors analyses revealed the basis for both ligand and DNA binding and also revealed receptor " gene regulation conformations representing both the activated and repressed states. Additionally, " metabolism crystallographic studies explained how NR LBD surfaces recognize discrete portions of transcriptional coregulators. The many structural snapshots of LBDs have also guided the development of synthetic ligands with therapeutic potential. Yet, the exclusive structural focus on isolated NR domains has made it difficult to conceptualize how all the NR polypeptide segments are coordinated physically and functionally in the context of receptor Journal of Molecular Endocrinology quaternary architectures. Newly emerged crystal structures of the peroxisome proliferator- activated receptor-g–retinoid X receptor a (PPARg–RXRa) heterodimer and hepatocyte nuclear factor (HNF)-4a homodimer have recently revealed the higher order organizations of these receptor complexes on DNA, as well as the complexity and uniqueness of their domain–domain interfaces.
    [Show full text]
  • 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.
    [Show full text]
  • Convergence of Multiple Mechanisms of Steroid Hormone Action
    Review 569 Convergence of Multiple Mechanisms of Steroid Hormone Action Authors S. K. Mani 1 * , P. G. Mermelstein 2 * , M. J. Tetel 3 * , G. Anesetti 4 * Affi liations 1 Department of Molecular & Cellular Biology and Neuroscience, Baylor College of Medicine, Houston, TX, USA 2 Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA 3 Neuroscience Program, Wellesley College, Wellesley, MA, USA 4 Departamento de Hostologia y Embriologia, Facultad de Medicine, Universidad de la Republica, Montevideo, Uruguay Key words Abstract receptors can also be activated in a “ligand-inde- ● ▶ estrogen ▼ pendent” manner by other factors including neu- ● ▶ progesterone Steroid hormones modulate a wide array of rotransmitters. Recent studies indicate that rapid, ▶ ● signaling physiological processes including development, nonclassical steroid eff ects involve extranuclear ● ▶ cross-talk metabolism, and reproduction in various species. steroid receptors located at the membrane, which ● ▶ ovary ● ▶ brain It is generally believed that these biological eff ects interact with cytoplasmic kinase signaling mol- are predominantly mediated by their binding to ecules and G-proteins. The current review deals specifi c intracellular receptors resulting in con- with various mechanisms that function together formational change, dimerization, and recruit- in an integrated manner to promote hormone- ment of coregulators for transcription-dependent dependent actions on the central and sympathetic genomic actions (classical mechanism). In addi- nervous systems. tion, to their cognate ligands, intracellular steroid Abbreviations gene expression and function. Interestingly, not ▼ all the “classical” receptors are intranuclear and CBP CREB binding protein can be associated at the membrane. As described CRE CREB response element in this review, extranuclear ERs and PRs at the DAR Dopamine receptor (DAR) membrane or in the cytoplasm can interact with ER Estrogen receptor G proteins and signaling kinases, and other G received 13 .
    [Show full text]