Transcriptional Control of the Glucocorticoid Receptor: Cpg Islands, Epigenetics and More

Total Page:16

File Type:pdf, Size:1020Kb

Transcriptional Control of the Glucocorticoid Receptor: Cpg Islands, Epigenetics and More Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more. Jonathan D. Turner, Simone R. Alt, Lei Cao, Sara Vernocchi, Slavena Trifonova, Nadia Battello, Claude P. Muller To cite this version: Jonathan D. Turner, Simone R. Alt, Lei Cao, Sara Vernocchi, Slavena Trifonova, et al.. Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more.. Biochemical Pharmacology, Elsevier, 2010, 80 (12), pp.1860. 10.1016/j.bcp.2010.06.037. hal-00637161 HAL Id: hal-00637161 https://hal.archives-ouvertes.fr/hal-00637161 Submitted on 31 Oct 2011 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Accepted Manuscript Title: Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more. Authors: Jonathan D. Turner, Simone R. Alt, Lei Cao, Sara Vernocchi, Slavena Trifonova, Nadia Battello, Claude P. Muller PII: S0006-2952(10)00471-5 DOI: doi:10.1016/j.bcp.2010.06.037 Reference: BCP 10618 To appear in: BCP Received date: 4-5-2010 Revised date: 18-6-2010 Accepted date: 21-6-2010 Please cite this article as: Turner JD, Alt SR, Cao L, Vernocchi S, Trifonova S, Battello N, Muller CP, Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more., Biochemical Pharmacology (2010), doi:10.1016/j.bcp.2010.06.037 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. 1 GR Transcription 2 3 4 5 1 Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more. 6 7 8 2 9 10 1,2 1,2 1,2 1,2 11 3 Jonathan D. Turner , Simone R. Alt , Lei Cao , Sara Vernocchi , Slavena 12 13 4 Trifonova1,2, Nadia Battello1, Claude P. Muller1,2 14 15 16 5 17 18 19 6 1Institute of Immunology, CRP-Santé / Laboratoire National de Santé, 20A rue Auguste 20 21 22 7 Lumière, L-1950, Luxembourg 23 24 25 8 2Department of Immunology, Graduate School of Psychobiology, University of 26 27 28 9 Trier, D-54290, Germany 29 30 10 31 32 33 11 34 35 36 12 37 38 39 13 Running title: GR transcription 40 41 42 14 43 44 45 15 46 47 48 16 Address for correspondence: 49 50 17 Claude P. Muller,Accepted Institute of Immunology, CRP -Santé/LaboratoireManuscript National de Santé, 51 52 18 20A rue Auguste Lumière, L-1950 Grand Duchy of Luxembourg. 53 19 Tel: +352 490604 Fax: +352 490686 54 55 20 Email: [email protected] 56 21 57 58 59 60 61 62 63 1 64 Page 1 of 32 65 1 GR Transcription 2 3 4 1 Abstract 5 6 2 The unique variability in the 5’ region of the GR gene, with 9 alternative first exons and 7 8 9 3 13 splice variants plays a critical role in transcriptional control maintaining homeostasis 10 11 4 of the glucocorticoid receptor (GR). This 5’mRNA heterogeneity, common to all species 12 13 5 investigated, remains untranslated since the alternative first exons are spliced to exon 2 14 15 16 6 immediately upstream of the translation initiation codon. These alternative first exons are 17 18 7 located either immediately upstream of the coding exons in the CpG island (exons B-H 19 20 21 8 and J), or further upstream (exons 1A and 1I). The mechanisms regulating the differential 22 23 9 usage of these first exons in different tissues and individuals, and the role of the 5’UTR 24 25 26 10 in the splicing of the coding exons are still poorly understood. Here we review some of 27 28 11 the mechanisms that have so far been identified. Data from our laboratory and others 29 30 12 have shown that the multiple first exons represent only a first layer of complexity 31 32 33 13 orchestrated probably by tissue-specific transcription factors. Modulation of alternative 34 35 14 first exon activity by epigenetic methylation of their promoters represents a second layer 36 37 38 15 of complexity at least partially controlled by perinatal programming. The alternative 39 40 16 promoter usage also appears to affect the 3’ splicing generating the different GR coding 41 42 43 17 variants, GR, GRβ, and GR-P. Aberrant GR levels are associated with stress-related 44 45 18 disorders such as depression, and affect social behaviour, mood, learning and memory. 46 47 48 19 Dissecting how tissue-specific GR levels are regulated, in particular in the brain, is a first 49 50 20 step to understandAccepted the significance of aberrant GRManuscript levels in disease and behaviour. 51 52 53 21 54 55 56 22 Keywords: Glucocorticoid receptor; Promoter; Methylation; Epigenetic variability; 57 58 23 alternative exon splicing 59 60 61 24 62 63 2 64 Page 2 of 32 65 1 GR Transcription 2 3 4 1 Introduction 5 6 7 2 Stress causes an increasing disease burden at least in Western societies. Economic losses 8 9 3 due to stress may be as high as 3-4% of the European gross national product. In addition, 10 11 12 4 stress has detrimental effects on behaviour, mood, learning and memory that are difficult 13 14 5 to quantify. Glucocorticoids (GCs), the key stress mediators, exert profound effects on a 15 16 6 wide range of physiological and developmental processes that are crucial for the 17 18 19 7 adaptation to stress. Psychosocial stress has also been implicated in the development of 20 21 8 mental disorders, including schizophrenia, anxiety disorders, and depression. 22 23 24 9 GCs act via binding to two types of intracellular nuclear receptors, i.e. the glucocorticoid 25 26 10 receptor (GR) and the mineralocorticoid receptor (MR). Upon binding of GCs, these 27 28 29 11 receptors translocate to the nucleus where they regulate the activity of specific target 30 31 12 genes, in a cell-type specific manner, as transcription factors. This review focuses on the 32 33 34 13 classical GR, NR3C1. Numerous factors have been demonstrated to affect the 35 36 14 responsiveness to GC by regulating GR activity, such as GR co-activators and co- 37 38 15 repressors [1], GR splice-variants [2-4], and GR isoforms [5-7]. In addition, and perhaps 39 40 41 16 most important for GC responsiveness is the expression level of GR protein [8-11], 42 43 17 determined by the mRNA level. 44 45 46 18 In eukaryotic cells, gene expression is controlled by a variety of mechanisms, both at 47 48 19 transcriptional and translational levels, including chromatin condensation, transcription 49 50 Accepted Manuscript 51 20 initiation, DNA methylation, alternative RNA splicing, mRNA stability and others. The 52 53 21 GR is an ubiquitously expressed nuclear hormone receptor, however, levels of both 54 55 22 mRNA and protein vary widely between cell and tissue types. Over the last few years we 56 57 58 23 and others have contributed to the significant progress that has been made to unravel the 59 60 61 62 63 3 64 Page 3 of 32 65 1 GR Transcription 2 3 4 1 transcriptional mechanisms determining the tissue specific control of GR levels, that will 5 6 7 2 be reviewed here. 8 9 3 10 11 12 4 Structure of the NR3C1 gene 13 14 5 The human GR gene (OMIM + 138040; NR3C1) is located on chromosome 5q31-q32 15 16 6 [12] and contains 8 translated exons (2-9) and 9 untranslated alternative first exons. We 17 18 19 7 and others have shown that GR levels are under the transcriptional control of a complex 20 21 8 5’ structure of the gene, containing the untranslated first exons important for differential 22 23 24 9 expression of the GR. All of the alternative first exons identified are located in one of the 25 26 10 two promoter regions: the proximal or the distal promoter region, located approximately 27 28 29 11 5kb and 30kb upstream of the translation start site, respectively [13-18]. Alternative first 30 31 12 exons 1A, and 1I are under the control of promoters in the distal promoter region, 32 33 34 13 whereas the promoters of exons 1D, 1J, 1E, 1B, 1F, 1C (1C1-3), 1H (Fig. 1A) are located 35 36 14 in the proximal promoter region [19, 20]. Exons 1D to 1H are found in an upstream CpG 37 38 15 island with a high sequence homology between rats and humans. 39 40 41 16 The region- or tissue-specific usage of alternative first exons leading to different GR 42 43 17 mRNA transcripts [19, 20] (Fig. 1 B) provides a mechanism for the local fine-tuning of 44 45 46 18 GR levels. Since the ATG start codon lies only in the common exon 2, this 5’mRNA 47 48 19 heterogeneity remains untranslated, but is important for translational regulation [21].
Recommended publications
  • TGF-Β1 Signaling Targets Metastasis-Associated Protein 1, a New Effector in Epithelial Cells
    Oncogene (2011) 30, 2230–2241 & 2011 Macmillan Publishers Limited All rights reserved 0950-9232/11 www.nature.com/onc ORIGINAL ARTICLE TGF-b1 signaling targets metastasis-associated protein 1, a new effector in epithelial cells SB Pakala1, K Singh1,3, SDN Reddy1, K Ohshiro1, D-Q Li1, L Mishra2 and R Kumar1 1Department of Biochemistry and Molecular Biology and Institute of Coregulator Biology, The George Washington University Medical Center, Washington, DC, USA and 2Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA In spite of a large number of transforming growth factor b1 gene chromatin in response to upstream signals. The (TGF-b1)-regulated genes, the nature of its targets with TGF-b1-signaling is largely mediated by Smad proteins roles in transformation continues to be poorly understood. (Massague et al., 2005) where Smad2 and Smad3 are Here, we discovered that TGF-b1 stimulates transcription phosphorylated by TGF-b1-receptors and associate with of metastasis-associated protein 1 (MTA1), a dual master the common mediator Smad4, which translocates to the coregulator, in epithelial cells, and that MTA1 status is a nucleus to participate in the expression of TGF-b1-target determinant of TGF-b1-induced epithelial-to-mesenchymal genes (Deckers et al., 2006). Previous studies have shown transition (EMT) phenotypes. In addition, we found that that CUTL1, also known as CDP (CCAAT displacement MTA1/polymerase II/activator protein-1 (AP-1) co-activator protein), a target of TGF-b1, is needed for its short-term complex interacts with the FosB-gene chromatin and stimu- effects of TGF-b1 on cell motility involving Smad4- lates its transcription, and FosB in turn, utilizes FosB/histone dependent pathway (Michl et al.,2005).
    [Show full text]
  • Conditional Deletion of HIF-1Α Provides New Insight
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.16.426940; this version posted January 16, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Conditional deletion of HIF-1 provides new insight regarding the murine response to gastrointestinal infection with Salmonella Typhimurium Laura Robrahn *, Aline Dupont †, Sandra Jumpertz *, Kaiyi Zhang †, Christian H. Holland ‡ §, Joël Guillaume ¶1, Sabrina Rappold *, Vuk Cerovic ¶, Julio Saez-Rodriguez ‡ §, Mathias W. Hornef † || #, and Thorsten Cramer * ** †† ‡‡ * Department of General, Visceral and Transplantation Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany; † Institute of Medical Microbiology, RWTH University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany; ‡ Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University, and Heidelberg University Hospital, Bioquant, Heidelberg, Germany; § Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, Aachen, Germany; ¶ Institute of Molecular Medicine, RWTH University Hospital, Aachen, Germany; ** ESCAM – European Surgery Center Aachen Maastricht, Germany and The Netherlands; †† Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands; ‡‡ NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht,
    [Show full text]
  • Rbpj-Dependent and -Independent Notch2 Signaling Regulates
    RBPJ-DEPENDENT AND -INDEPENDENT NOTCH2 SIGNALING REGULATES CILIARY BODY DEVELOPMENT IN THE MOUSE EYE By Yi Zhou B.S., Tsinghua University, 2010 Submitted to the graduate degree program in Anatomy and Cell Biology and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. ________________________________ Ting Xie, Ph.D., Co-Chair ________________________________ William Kinsey, Ph.D., Co-Chair ________________________________ Dale Abrahamson, Ph.D. ________________________________ Peter Smith, Ph.D. ________________________________ Jerry Workman, Ph.D. Date Defended: Jan 6th, 2016 The Dissertation Committee for Yi Zhou certifies that this is the approved version of the following dissertation: RBPJ-DEPENDENT AND -INDEPENDENT NOTCH2 SIGNALING REGULATES CILIARY BODY DEVELOPMENT IN THE MOUSE EYE ________________________________ Ting Xie, Ph.D., Co-Chair ________________________________ William Kinsey, Ph.D., Co-Chair Date Approved: Jan 15th, 2016 ii ABSTRACT The ciliary body (CB) is a two-layered structure in the anterior eye, which is composed of the pigmented outer ciliary epithelium (OCE) and the non-pigmented inner ciliary epithelium (ICE). It is responsible for aqueous humor secretion and lens accommodation. Despite the important roles in maintaining normal eye functions, its development still remains poorly understood. The Notch signaling pathway is an evolutionarily conserved pathway that has diverse functions during tissue development and homeostasis. Canonical Notch signaling is mediated through the recombination signal binding protein for immunoglobulin kappa J region (RBPJ)-dependent transcription activation and repression. In this study, I have demonstrated that Notch2 and RBPJ are important regulators of CB development by conditionally deleting them in the developing CB.
    [Show full text]
  • Estradiol Inhibits ER Stress-Induced Apoptosis Through
    Laboratory Investigation (2014) 94, 906–916 & 2014 USCAP, Inc All rights reserved 0023-6837/14 17b-Estradiol inhibits ER stress-induced apoptosis through promotion of TFII-I-dependent Grp78 induction in osteoblasts Yun-Shan Guo1,2,5, Zhen Sun1,5, Jie Ma3,5, Wei Cui4,5, Bo Gao1, Hong-Yang Zhang1, Yue-Hu Han1, Hui-Min Hu1, Long Wang1, Jing Fan1, Liu Yang1, Juan Tang2 and Zhuo-Jing Luo1 Although many studies have suggested that estrogen prevents postmenopausal bone loss partially due to its anti-apoptosis effects in osteoblasts, the underlying mechanism has not been fully elucidated. In the present study, we found that 17b-estradiol (17b-E2), one of the primary estrogens, inhibited endoplasmic reticulum (ER) stress-induced apoptosis in MC3T3-E1 cells and primary osteoblasts. Interestingly, 17b-E2-promoted Grp78 induction, but not CHOP induction in response to ER stress. We further confirmed that Grp78-specific siRNA reversed the inhibition of 17b-E2 on ER stress-induced apoptosis by activating caspase-12 and caspase-3. Moreover, we found that 17b-E2 markedly increased the phosphorylated TFII-I levels and nuclear localization of TFII-I in ER stress conditions. 17b-E2 stimulated Grp78 promoter activity in a dose-dependent manner in the presence of TFII-I and enhanced the binding of TFII-I to the Grp78 promoter. In addition, 17b-E2 notably increased phosphorylated ERK1/2 levels and Ras kinase activity in MC3T3-E1 cells. The ERK1/2 activity-specific inhibitor U0126 remarkably blocked 17b-E2-induced TFII-I phosphorylation and Grp78 expression in response to ER stress. Together, 17b-E2 protected MC3T3-E1 cells against ER stress-induced apoptosis by promoting Ras-ERK1/2-TFII-I signaling pathway-dependent Grp78 induction.
    [Show full text]
  • In Vitro Targeting of Transcription Factors to Control the Cytokine Release Syndrome in 2 COVID-19 3
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.29.424728; this version posted December 30, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 1 In vitro Targeting of Transcription Factors to Control the Cytokine Release Syndrome in 2 COVID-19 3 4 Clarissa S. Santoso1, Zhaorong Li2, Jaice T. Rottenberg1, Xing Liu1, Vivian X. Shen1, Juan I. 5 Fuxman Bass1,2 6 7 1Department of Biology, Boston University, Boston, MA 02215, USA; 2Bioinformatics Program, 8 Boston University, Boston, MA 02215, USA 9 10 Corresponding author: 11 Juan I. Fuxman Bass 12 Boston University 13 5 Cummington Mall 14 Boston, MA 02215 15 Email: [email protected] 16 Phone: 617-353-2448 17 18 Classification: Biological Sciences 19 20 Keywords: COVID-19, cytokine release syndrome, cytokine storm, drug repurposing, 21 transcriptional regulators 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.12.29.424728; this version posted December 30, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 22 Abstract 23 Treatment of the cytokine release syndrome (CRS) has become an important part of rescuing 24 hospitalized COVID-19 patients. Here, we systematically explored the transcriptional regulators 25 of inflammatory cytokines involved in the COVID-19 CRS to identify candidate transcription 26 factors (TFs) for therapeutic targeting using approved drugs.
    [Show full text]
  • Enhancer, Hs3 Igh ′ Activator of the Murine 3 Yin Yang 1 Is A
    Yin Yang 1 Is a Lipopolysaccharide-Inducible Activator of the Murine 3 ′ Igh Enhancer, hs3 This information is current as Steven J. Gordon, Shireen Saleque and Barbara K. Birshtein of September 23, 2021. J Immunol 2003; 170:5549-5557; ; doi: 10.4049/jimmunol.170.11.5549 http://www.jimmunol.org/content/170/11/5549 Downloaded from References This article cites 66 articles, 40 of which you can access for free at: http://www.jimmunol.org/content/170/11/5549.full#ref-list-1 Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average by guest on September 23, 2021 Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2003 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Yin Yang 1 Is a Lipopolysaccharide-Inducible Activator of the Murine 3؅ Igh Enhancer, hs31 Steven J. Gordon, Shireen Saleque,2 and Barbara K. Birshtein3 The 3؅ Igh enhancers, DNase I hypersensitive site (hs) 3B and/or hs4, are required for germline transcription, and hence, class switch recombination for multiple isotypes.
    [Show full text]
  • Endothelial Specific YY1 Deletion Restricts Tumor Angiogenesis And
    www.nature.com/scientificreports OPEN Endothelial specifc YY1 deletion restricts tumor angiogenesis and tumor growth Huan Liu1,2, Yikai Qiu1, Xiuying Pei1, Ramamurthy Chitteti2, Rebbeca Steiner2, Shuya Zhang1,2* & Zheng Gen Jin2* Angiogenesis is a physiological process for the formation of new blood vessels from the pre-existing vessels and it has a vital role in the survival and growth of neoplasms. During tumor angiogenesis, the activation of the gene transcriptions in vascular endothelial cells (ECs) plays an essential role in the promotion of EC proliferation, migration, and vascular network development. However, the molecular mechanisms underlying transcriptional regulation of EC and tumor angiogenesis remains to be fully elucidated. Here we report that the transcription factor Yin Yang 1 (YY1) in ECs is critically involved in tumor angiogenesis. First, we utilized a tamoxifen-inducible EC-specifc YY1 defcient mouse model and showed that YY1 deletion in ECs inhibited the tumor growth and tumor angiogenesis. Using the in vivo matrigel plug assay, we then found that EC-specifc YY1 ablation inhibited growth factor-induced angiogenesis. Furthermore, vascular endothelial growth factor (VEGF)-induced EC migration was diminished in YY1-depleted human umbilical vein endothelial cells (HUVECs). Finally, a rescue experiment revealed that YY1-regulated BMP6 expression in ECs was involved in EC migration. Collectively, our results demonstrate that endothelial YY1 has a crucial role in tumor angiogenesis and suggest that targeting endothelial
    [Show full text]
  • The YY1 Transcription Factor Is a Component of Ribonucleoprotein Complexes In
    The YY1 Transcription Factor is a Component of Ribonucleoprotein Complexes in Xenopus laevis Oocytes and Embryos. A Thesis Submitted to the College of Graduate Studies and Research In Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy In the Department of Anatomy and Cell Biology University of Saskatchewan Saskatoon Saskatchewan By Andrew Douglas Ficzycz Spring 2003 © Copyright Andrew Do uglas Ficzyc z, 2003. All rights reserv ed. Permission to use P ERMIS SION TO US E I n presenting this thesis in partial f ulfilment of the r equirements for a P ostgraduate degree f rom the University of S askatchew an, I agree that the Libraries of this U nivers ity may make it f reely available f or inspection. I f urther agree that permission f or copying of this thesis in any manner, in w hole or in part, for s cholarly purpos es may be granted by the professor or profess ors w ho supervis ed my thesis w ork or , in their absence, by the Head of the D epartment or the D ean of the College in w hich my thesis work was done. I t is unders tood that any copying or publication or use of this thesis or parts thereof f or financial gain s hall not be allowed without my w ritten permis sion. It is also understood that due r ecognition shall be given to me and to the Univer s ity of Saskatchew an in any scholarly use w hich may be made of any material in my thesis.
    [Show full text]
  • KRAS/NF-Kb/YY1/Mir-489 Signaling Axis Controls Pancreatic Cancer
    Published OnlineFirst October 28, 2016; DOI: 10.1158/0008-5472.CAN-16-1898 Cancer Molecular and Cellular Pathobiology Research KRAS/NF-kB/YY1/miR-489 Signaling Axis Controls Pancreatic Cancer Metastasis Peng Yuan1,2, Xiao-Hong He1,2, Ye-Fei Rong3, Jing Cao4, Yong Li5, Yun-Ping Hu6, Yingbin Liu6, Dangsheng Li7, Wenhui Lou3, and Mo-Fang Liu1,2,4 Abstract KRAS activation occurring in more than 90% of pancreatic inhibited migration and metastasis by targeting the extracellular ductal adenocarcinomas (PDAC) drives progression and metas- matrix factors ADAM9 and MMP7. miR-489 downregulation tasis, but the underlying mechanisms involved in these processes elevated levels of ADAM9 and MMP7, thereby enhancing are still poorly understood. Here, we show how KRAS acts through the migration and metastasis of PDAC cells. Together, our inflammatory NF-kB signaling to activate the transcription factor results establish a pivotal mechanism of PDAC metastasis and YY1, which represses expression of the tumor suppressor gene suggest miR-489 as a candidate therapeutic target for their attack. miR-489. In PDAC cells, repression of miR-489 by KRAS signaling Cancer Res; 77(1); 100–11. Ó2016 AACR. KRAS in pancreatic tumors, the most common of which is Introduction G12D KRAS (4). Mounting evidence indicates that such onco- Pancreatic cancer (PDAC) is one of the most lethal malignant genic mutations play critical roles in both the initiation and the tumors, with a 5-year survival rate less than 8% from 2005 to 2011 progression of pancreatic cancer via persistent activation of in United States (1). To date, lack of effective screening tool to KRAS signaling pathways (5).
    [Show full text]
  • Relief of YY1 Transcriptional Repression by Adenovirus E1A Is Mediated by E1A-Associated Protein P300
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press Relief of YY1 transcriptional repression by adenovirus E1A is mediated by E1A-associated protein p300 Jeng-Shin Lee, 1"2 Katherine M. Galvin, l's Raymond H. See, l's Richard Eckner, 3 David Livingston, 3 Elizabeth Moran, 4 and Yang Shi 1'2'6 ~Department of Pathology, 2Committee on Virology, Harvard Medical School, Boston, Massachusetts 02115 USA; aDivision of Cellular and Molecular Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115 USA; 4The Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 USA YY1 represses transcription when bound upstream of transcriptional initiation sites. This repression can be relieved by adenovirus E1A. Here, we present genetic evidence that the ability of E1A to relieve YY1 repression was impaired by mutations that affect EIA binding to its associated protein p300. This suggests that E1A may modulate the repressor activity of YY1 by binding to p300, which may be physically complexed with YY1. A YY1/p300 protein complex in vivo was demonstrated by several independent approaches, and the YYl-interacting domain was mapped to the carboxy-terminal region of p300, distinct from the E1A-binding site. Unlike E2F/RB, the YY1/p300 complex is not disrupted by E1A. Functional studies using recombinant p300 demonstrated unequivocally that p300 is capable of mediating E1A-induced transcriptional activation through YY1. Taken together, these results reveal, for the first time, a YY1/p300 complex that is targeted by EIA and demonstrate a function for p300 in mediating interactions between YY1 and E1A.
    [Show full text]
  • PDF-Document
    Supplementary Material Investigating the role of microRNA and Transcription Factor co-regulatory networks in Multiple Sclerosis pathogenesis Nicoletta Nuzziello1, Laura Vilardo2, Paride Pelucchi2, Arianna Consiglio1, Sabino Liuni1, Maria Trojano3 and Maria Liguori1* 1National Research Council, Institute of Biomedical Technologies, Bari Unit, Bari, Italy 2National Research Council, Institute of Biomedical Technologies, Segrate Unit, Milan, Italy 3Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy Supplementary Figure S1 Frequencies of GO terms and canonical pathways. (a) Histogram illustrates the GO terms associated to assembled sub-networks. (b) Histogram illustrates the canonical pathways associated to assembled sub-network. a b Legends for Supplementary Tables Supplementary Table S1 List of feedback (FBL) and feed-forward (FFL) loops in miRNA-TF co-regulatory network. Supplementary Table S2 List of significantly (adj p-value < 0.05) GO-term involved in MS. The first column (from the left) listed the GO-term (biological processes) involved in MS. For each functional class, the main attributes (gene count, p-value, adjusted p-value of the enriched terms for multiple testing using the Benjamini correction) have been detailed. In the last column (on the right), we summarized the target genes involved in each enriched GO-term. Supplementary Table S3 List of significantly (adj p-value < 0.05) enriched pathway involved in MS. The first column (from the left) listed the enriched pathway involved in MS. For each pathway, the main attributes (gene count, p-value, adjusted p-value of the enriched terms for multiple testing using the Benjamini correction) have been detailed. In the last column (on the right), we summarized the target genes involved in each enriched pathway.
    [Show full text]
  • YY1 Suppresses FEN1 Over-Expression and Drug
    Wang et al. BMC Cancer (2015) 15:50 DOI 10.1186/s12885-015-1043-1 RESEARCH ARTICLE Open Access YY1 suppresses FEN1 over-expression and drug resistance in breast cancer Jianwei Wang1, Lina Zhou1,2, Zhi Li3, Ting Zhang1, Wenpeng Liu1, Zheng Liu2, Yate-Ching Yuan2, Fan Su2,LuXu3, Yan Wang3, Xiaotong Zhou3, Hong Xu4, Yuejin Hua4, Ying-Jie Wang5, Li Zheng2, Yue-E Teng3* and Binghui Shen2* Abstract Background: Drug resistance is a major challenge in cancer therapeutics. Abundant evidence indicates that DNA repair systems are enhanced after repetitive chemotherapeutic treatments, rendering cancers cells drug-resistant. Flap endonuclease 1 (FEN1) plays critical roles in DNA replication and repair and in counteracting replication stress, which is a key mechanism for many chemotherapeutic drugs to kill cancer cells. FEN1 was previously shown to be upregulated in response to DNA damaging agents. However, it is unclear about the transcription factors that regulate FEN1 expression in human cancer. More importantly, it is unknown whether up-regulation of FEN1 has an adverse impact on the prognosis of chemotherapeutic treatments of human cancers. Methods: To reveal regulation mechanism of FEN1 expression, we search and identify FEN1 transcription factors or repressors and investigate their function on FEN1 expression by using a combination of biochemical, molecular, and cellular approaches. Furthermore, to gain insights into the impact of FEN1 levels on the response of human cancer to therapeutic treatments, we determine FEN1 levels in human breast cancer specimens and correlate them to the response to treatments and the survivorship of corresponding breast cancer patients. Results: We observe that FEN1 is significantly up-regulated upon treatment of chemotherapeutic drugs such as mitomycin C (MMC) and Taxol in breast cancer cells.
    [Show full text]