1 Mechanistic Analysis of Enhancer Sequences in the Estrogen Receptor

Total Page:16

File Type:pdf, Size:1020Kb

1 Mechanistic Analysis of Enhancer Sequences in the Estrogen Receptor bioRxiv preprint doi: https://doi.org/10.1101/2020.11.08.373555; this version posted November 9, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. 1 Mechanistic analysis of enhancer sequences in the Estrogen Receptor 2 transcriptional program 3 Shayan Tabe-Bordbar 4 Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, 5 United States of America. Email: [email protected] 6 You Jin Song 7 Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, 8 Urbana, IL, United States of America, Email: [email protected] 9 Bryan J. Lunt 10 Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, 11 United States of America. Email: [email protected] 12 Kannanganattu V. Prasanth 13 Department of Cell and Developmental Biology, Cancer Center of Illinois, University of Illinois at 14 Urbana-Champaign, Urbana, IL, United States of America. Email: [email protected] 15 Saurabh Sinha 16 Department of Computer Science, Carl R. Woese Institute for Genomic Biology, Cancer Center 17 of Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America. 18 Email: [email protected] 19 Corresponding Author: 20 Saurabh Sinha 21 2122 Siebel Center, 201 N. Goodwin Ave, Urbana, IL 61801. USA. 22 Phone: 217-333-3233 23 Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.11.08.373555; this version posted November 9, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. 24 Abstract 25 Background: Estrogen Receptor α (ERα) is a major lineage determining transcription factor 26 (TF) in mammary gland development, orchestrating the expression of thousands of genes. 27 Dysregulation of ERα-mediated transcriptional program results in abnormal cell proliferation and 28 cancer. Transcriptomic and epigenomic profiling of breast cancer cell lines has revealed large 29 numbers of enhancers involved in this regulatory program, but how these enhancers encode 30 function in their sequence remains poorly understood. 31 Results: A subset of ERα-bound enhancers are transcribed into short bidirectional RNA 32 (enhancer RNA or eRNA), and this property is believed to be a reliable marker of active 33 enhancers. We therefore analyze thousands of ERα-bound enhancers and build quantitative, 34 mechanism-aware models to discriminate eRNAs from non-transcribing enhancers based on 35 their sequence. Our thermodynamics-based models provide insights into the roles of specific 36 TFs in ERα-mediated transcriptional program, many of which are supported by the literature. 37 We use in silico perturbations to predict TF-enhancer regulatory relationships and integrate 38 these findings with experimentally determined enhancer-promoter interactions to construct a 39 gene regulatory network. We also demonstrate that the model can prioritize breast cancer- 40 related sequence variants while providing mechanistic explanations for their function. Finally, we 41 experimentally validate the model-proposed mechanisms underlying three such variants. 42 Conclusions: We modeled the sequence-to-expression relationship in ERα-driven enhancers 43 and gained mechanistic insights into the workings of a major transcriptional program. Our model 44 is consistent with the current body of knowledge and its predictions are confirmed by 45 experimental observations. We believe this to be a promising approach to analysis of regulatory 46 sequences and variants. 47 Keywords 2 bioRxiv preprint doi: https://doi.org/10.1101/2020.11.08.373555; this version posted November 9, 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. 48 enhancers, gene regulatory network, eRNA, sequence-to-expression modeling, variant impact 49 prediction, thermodynamics-based modeling, breast cancer 50 51 Background 52 Breast cancer is the second most common cancer in women in the United States, with about 53 12.4% of women diagnosed with breast cancer during their lifetime (1). The most common 54 breast cancer subtype is characterized by increased activity of Estrogen Receptor α (ERα), a 55 protein that is activated by estrogen and in turn changes the expression of hundreds of genes. 56 Acting as a transcription factor (TF), it binds to thousands of DNA locations and influences the 57 expression levels of nearby genes (2). This cascade of events is called the “ERα transcriptional 58 program”, and aberrations in this program lead to increased cell proliferation and cancer. 59 Several drugs have been developed to treat the ER+ subtype of breast cancer by reversing 60 aberrations in the ERα program or interfering with its cancer-causing effects. However, about 61 50% of treated patients either do not respond to or develop acquired resistance against these 62 drugs (3). As such, there is great interest in characterizing the major principles and crucial 63 details of the ERα transcriptional program. 64 The regulatory information controlling gene expression programs are known to be in part stored 65 in relatively short stretches of DNA (between 1-2 kbp) called enhancers (4). Enhancers typically 66 harbor several binding sites for one or more transcription factors (TFs). Each binding site is an 67 approximately 10 base long DNA sequence with relatively high binding affinity for its 68 corresponding TF. Active enhancers – marked by specific chromatin marks – regulate 69 expression of genes in their spatial proximity through binding to a specific set of TFs. Notably, 70 the spatial proximity of a gene-enhancer pair depends on the chromatin conformation and is not 71 necessarily captured by their genomic distance. In other words, genes can be regulated by 3 bioRxiv preprint doi: https://doi.org/10.1101/2020.11.08.373555; this version posted November 9, 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. 72 enhancers located virtually anywhere on the genome. ERα is known to mainly regulate 73 expression of genes through binding to distant enhancers, as only about 5% of its binding sites 74 are located within a distance of 5 kb from Transcription Start Site (TSS) of any gene (5). 75 Over the past decade, biotechnological advancements such as RNA-sequencing (RNA-Seq) 76 and chromatin immunoprecipitation coupled with sequencing (ChIP-Seq) have dramatically 77 expanded our knowledge about the ERα transcriptional program. RNA-Seq assays provide data 78 on expression levels of all genes in different experimental conditions, while ChIP-seq 79 experiments reveal all the locations in the genome where ERα is bound in those conditions. Due 80 to its biological importance and potential impact, the ERα transcriptional program has been 81 studied not only through mainstream experimental approaches such as RNA-seq, ChIP seq, 82 and DNase-Seq (6,7), but also relatively more advanced and expensive experimental 83 techniques such as Chromatin Interaction Analysis by Paired-End Tag Sequencing (ChIA-PET) 84 (8,9), Global run-on sequencing (GRO-Seq) (10–13), and high-resolution chromatin 85 conformation capture (Hi-C) (14), resulting in an unprecedented wealth of diverse data 86 describing this system. 87 Here, we aimed to comprehensively characterize gene regulatory relationships in this system 88 through modeling of gene expression data, taking advantage of several of the above-mentioned 89 multi-omics data. A common approach to reconstruction of gene regulatory networks (GRN) is 90 to train a quantitative model that correctly predicts gene expression levels, and then interpret 91 the parameters of the model to identify regulatory relationships. There exist two major variations 92 of the gene expression prediction problem. In the “sequence-to-expression” formulation, DNA 93 sequences (enhancers) associated with a gene should bear the “footprints” of the TFs that 94 ostensibly regulate the gene, since a TF needs to physically bind to enhancers in order to 95 regulate the gene (Additional File 1: Figure S1). In the alternative, less constrained “expression- 96 to-expression” formulation, one attempts to relate a gene’s expression to the expression of its 4 bioRxiv preprint doi: https://doi.org/10.1101/2020.11.08.373555; this version posted November 9, 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. 97 potential regulators, without taking DNA sequence information into account. In the current study, 98 we considered the former formulation of the expression prediction problem in order to study the 99 ERα transcriptional program, reconstruct the underlying GRN, and decipher the sequence-level 100 encoding of the network. 101 Sequence-to-expression modeling in mammalian systems poses two key challenges: enhancer- 102 gene assignment, and enhancer-readout prediction. In enhancer-gene assignment, one must 103 identify the genomic location of enhancers regulating a gene’s expression and their relative 104 contributions in a particular condition. On the other hand, enhancer-readout prediction amounts 105 to quantifying the expression driven by a particular
Recommended publications
  • Functions of the Mineralocorticoid Receptor in the Hippocampus By
    Functions of the Mineralocorticoid Receptor in the Hippocampus by Aaron M. Rozeboom A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Cellular and Molecular Biology) in The University of Michigan 2008 Doctoral Committee: Professor Audrey F. Seasholtz, Chair Professor Elizabeth A. Young Professor Ronald Jay Koenig Associate Professor Gary D. Hammer Assistant Professor Jorge A. Iniguez-Lluhi Acknowledgements There are more people than I can possibly name here that I need to thank who have helped me throughout the process of writing this thesis. The first and foremost person on this list is my mentor, Audrey Seasholtz. Between working in her laboratory as a research assistant and continuing my training as a graduate student, I spent 9 years in Audrey’s laboratory and it would be no exaggeration to say that almost everything I have learned regarding scientific research has come from her. Audrey’s boundless enthusiasm, great patience, and eager desire to teach students has made my time in her laboratory a richly rewarding experience. I cannot speak of Audrey’s laboratory without also including all the past and present members, many of whom were/are not just lab-mates but also good friends. I also need to thank all the members of my committee, an amazing group of people whose scientific prowess combined with their open-mindedness allowed me to explore a wide variety of interests while maintaining intense scientific rigor. Outside of Audrey’s laboratory, there have been many people in Ann Arbor without whom I would most assuredly have gone crazy.
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Identification of NFIB As Recurrent Translocation Partner Gene of HMGIC in Pleomorphic Adenomas
    Oncogene (1998) 16, 865 ± 872 1998 Stockton Press All rights reserved 0950 ± 9232/98 $12.00 Identi®cation of NFIB as recurrent translocation partner gene of HMGIC in pleomorphic adenomas Jan MW Geurts1, Eric FPM Schoenmakers1, Eva RoÈ ijer2, Anna-Karin AstroÈ m2,GoÈran Stenman2 and Wim JM van de Ven1 1Laboratory for Molecular Oncology, Center for Human Genetics, University of Leuven & Flanders Interuniversity Institute for Biotechnology, B-3000 Leuven, Belgium; 2Laboratory of Cancer Genetics, Department of Pathology, GoÈteborg University, Sahlgrenska University Hospital, S-41345 GoÈteborg, Sweden Approximately 12% of all pleomorphic adenomas of the guished, i.e. tumors with rearrangements of 8q12 or salivary glands are characterized by chromosome 12q13 ± 15. Whereas the t(3;8)(p21;q12) is the most aberrations involving the chromosome segment 12q13 ± frequently observed translocation, many dierent 15. Several chromosomes have been found as transloca- chromosome segments can act as translocation tion partners of chromosome 12, and some of these partners of both 8q12 and 12q13 ± 15 (Sandros et al., recurrently. Recently, the HMGIC gene was identi®ed as 1990; Bullerdiek et al., 1993). The short arm of the target gene aected by the 12q13 ± 15 aberrations. chromosome 9, with breakpoints in the segment Here, we report the identi®cation and characterization of 9p12 ± 24, is the preferential translocation partner of a new translocation partner gene of HMGIC in chromosome 12q13 ± 15 (Stenman et al., 1994). The pleomorphic adenomas. 3'-RACE analysis of a primary reason for the variability of the breakpoints in 9p is adenoma with an apparently normal karyotype revealed likely to be due to the limited resolution of the banding an HMGIC fusion transcript containing ectopic se- pattern in 9p12 ± 24 and 12q13 ± 15, making it almost quences derived from the human NFIB gene, previously impossible to determine the exact locations of the mapped to chromosome band 9p24.1.
    [Show full text]
  • And EZH2-Regulated Gene, Has Differential Roles in AR-Dependent and -Independent Prostate Cancer
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No.4 RUNX1, an androgen- and EZH2-regulated gene, has differential roles in AR-dependent and -independent prostate cancer Ken-ichi Takayama1,2, Takashi Suzuki3, Shuichi Tsutsumi4, Tetsuya Fujimura5, Tomohiko Urano1,2, Satoru Takahashi6, Yukio Homma5, Hiroyuki Aburatani4 and Satoshi Inoue1,2,7 1 Department of Anti-Aging Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan 2 Department of Geriatric Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan 3 Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan 4 Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Meguro-ku, Tokyo, Japan 5 Department of Urology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan 6 Department of Urology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan 7 Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan Correspondence to: Satoshi Inoue, email: [email protected] Keywords: RUNX1, androgen receptor, EZH2, prostate cancer Received: October 02, 2014 Accepted: December 09, 2014 Published: December 10, 2014 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Androgen receptor (AR) signaling is essential for the development of prostate cancer. Here, we report that runt-related transcription factor (RUNX1) could be a key molecule for the androgen-dependence of prostate cancer. We found RUNX1 is a target of AR and regulated positively by androgen.
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 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. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 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. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Cutting Edge: Expression of IRF8 in Gastric Epithelial Cells Confers
    Cutting Edge: Expression of IRF8 in Gastric Epithelial Cells Confers Protective Innate Immunity against Helicobacter pylori Infection This information is current as of September 27, 2021. Ming Yan, Hongsheng Wang, Jiafang Sun, Wei Liao, Peng Li, Yin Zhu, Chengfu Xu, Jungsoo Joo, Yan Sun, Sadia Abbasi, Alexander Kovalchuk, Nonghua Lv, Warren J. Leonard and Herbert C. Morse III J Immunol published online 3 February 2016 Downloaded from http://www.jimmunol.org/content/early/2016/02/02/jimmun ol.1500766 http://www.jimmunol.org/ Supplementary http://www.jimmunol.org/content/suppl/2016/02/02/jimmunol.150076 Material 6.DCSupplemental Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on September 27, 2021 • Fast Publication! 4 weeks from acceptance to publication *average 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 © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published February 3, 2016, doi:10.4049/jimmunol.1500766 Th eJournal of Cutting Edge Immunology Cutting Edge: Expression of IRF8 in Gastric Epithelial Cells Confers Protective Innate Immunity against Helicobacter pylori Infection x x Ming Yan,*,1 Hongsheng Wang,†,1 Jiafang Sun,† Wei Liao,‡, Peng Li,‡, { † Yin Zhu, ChengfuXu,*JungsooJoo,*YanSun,*SadiaAbbasi,{ x Alexander Kovalchuk,† Nonghua Lv, Warren J.
    [Show full text]
  • 1714 Gene Comprehensive Cancer Panel Enriched for Clinically Actionable Genes with Additional Biologically Relevant Genes 400-500X Average Coverage on Tumor
    xO GENE PANEL 1714 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes 400-500x average coverage on tumor Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11 IL6R KAT5 ACVR2A
    [Show full text]
  • Diverse Signaling Pathways Modulate Nuclear Receptor Recruitment of N-Cor and SMRT Complexes (Estrogen Receptor͞tamoxifen͞corepressor Complex)
    Proc. Natl. Acad. Sci. USA Vol. 95, pp. 2920–2925, March 1998 Biochemistry Diverse signaling pathways modulate nuclear receptor recruitment of N-CoR and SMRT complexes (estrogen receptorytamoxifenycorepressor complex) ROBERT M. LAVINSKY*†,KRISTEN JEPSEN*†,THORSTEN HEINZEL*, JOSEPH TORCHIA*, TINA-MARIE MULLEN‡, RACHEL SCHIFF§,ALFONSO LEON DEL-RIO*, MERCEDES RICOTE¶,SALLY NGO¶,JOSLIN GEMSCH‡, SUSAN G. HILSENBECK§,C.KENT OSBORNE§,CHRISTOPHER K. GLASS¶,MICHAEL G. ROSENFELD*, \ AND DAVID W. ROSE‡ *Howard Hughes Medical Institute, ¶Cellular and Molecular Medicine, ‡Whittier Diabetes Program, and †UCSD Graduate Program in Biology, Department and School of Medicine, University of California at San Diego, La Jolla, CA 92093-0648; and §Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78284-7884 Contributed by Michael G. Rosenfeld, January 9, 1998 ABSTRACT Several lines of evidence indicate that the partial agonistic activity (31). Various agents that raise intra- nuclear receptor corepressor (N-CoR) complex imposes li- cellular cAMP levels or stimulate the rasyMAP kinase path- gand dependence on transcriptional activation by the retinoic way can similarly cause estrogen receptor (ER) activation in acid receptor and mediates the inhibitory effects of estrogen the presence of tamoxifen or the absence of any activating receptor antagonists, such as tamoxifen, suppressing a con- ligand (9, 32–35). In this manuscript we show that diverse stitutive N-terminal, Creb-binding proteinycoactivator com- molecular strategies regulate the association of N-CoR- or plex-dependent activation domain. Functional interactions SMRT-containing complexes with specific nuclear receptors, between specific receptors and N-CoR or SMRT corepressor including the nature of the ligand, the levels of available complexes are regulated, positively or negatively, by diverse N-CoRySMRT, and the action of diverse protein kinase- signal transduction pathways.
    [Show full text]
  • The Staurotypus Turtles and Aves Share the Same Origin of Sex Chromosomes but Evolved Different Types of Heterogametic Sex Determination
    The Staurotypus Turtles and Aves Share the Same Origin of Sex Chromosomes but Evolved Different Types of Heterogametic Sex Determination Taiki Kawagoshi1, Yoshinobu Uno1, Chizuko Nishida2, Yoichi Matsuda1,3* 1 Laboratory of Animal Genetics, Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan, 2 Department of Natural History Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan, 3 Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan Abstract Reptiles have a wide diversity of sex-determining mechanisms and types of sex chromosomes. Turtles exhibit temperature- dependent sex determination and genotypic sex determination, with male heterogametic (XX/XY) and female heterogametic (ZZ/ZW) sex chromosomes. Identification of sex chromosomes in many turtle species and their comparative genomic analysis are of great significance to understand the evolutionary processes of sex determination and sex chromosome differentiation in Testudines. The Mexican giant musk turtle (Staurotypus triporcatus, Kinosternidae, Testudines) and the giant musk turtle (Staurotypus salvinii) have heteromorphic XY sex chromosomes with a low degree of morphological differentiation; however, their origin and linkage group are still unknown. Cross-species chromosome painting with chromosome-specific DNA from Chinese soft-shelled turtle (Pelodiscus sinensis) revealed that the X and Y chromosomes of S. triporcatus have homology with P. sinensis chromosome 6, which corresponds to the chicken Z chromosome. We cloned cDNA fragments of S. triporcatus homologs of 16 chicken Z-linked genes and mapped them to S. triporcatus and S. salvinii chromosomes using fluorescence in situ hybridization. Sixteen genes were localized to the X and Y long arms in the same order in both species.
    [Show full text]
  • Mutational Landscape and Clinical Outcome of Patients with De Novo Acute Myeloid Leukemia and Rearrangements Involving 11Q23/KMT2A
    Mutational landscape and clinical outcome of patients with de novo acute myeloid leukemia and rearrangements involving 11q23/KMT2A Marius Billa,1,2, Krzysztof Mrózeka,1,2, Jessica Kohlschmidta,b, Ann-Kathrin Eisfelda,c, Christopher J. Walkera, Deedra Nicoleta,b, Dimitrios Papaioannoua, James S. Blachlya,c, Shelley Orwicka,c, Andrew J. Carrolld, Jonathan E. Kolitze, Bayard L. Powellf, Richard M. Stoneg, Albert de la Chapelleh,i,2, John C. Byrda,c, and Clara D. Bloomfielda,c aThe Ohio State University Comprehensive Cancer Center, Columbus, OH 43210; bAlliance for Clinical Trials in Oncology Statistics and Data Center, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210; cDivision of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210; dDepartment of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294; eNorthwell Health Cancer Institute, Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY 11042; fDepartment of Internal Medicine, Section on Hematology & Oncology, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157; gDepartment of Medical Oncology, Dana-Farber/Partners Cancer Care, Boston, MA 02215; hHuman Cancer Genetics Program, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210; and iDepartment of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210 Contributed by Albert de la Chapelle, August 28, 2020 (sent for review July 17, 2020; reviewed by Anne Hagemeijer and Stefan Klaus Bohlander) Balanced rearrangements involving the KMT2A gene, located at patterns that include high expression of HOXA genes and thereby 11q23, are among the most frequent chromosome aberrations in contribute to leukemogenesis (14–16).
    [Show full text]
  • Figure S17 Figure S16
    immune responseregulatingcellsurfacereceptorsignalingpathway ventricular cardiacmuscletissuedevelopment t cellactivationinvolvedinimmuneresponse intrinsic apoptoticsignalingpathway single organismalcelladhesion cholesterol biosyntheticprocess myeloid leukocytedifferentiation determination ofadultlifespan response tointerferongamma muscle organmorphogenesis endothelial celldifferentiation brown fatcelldifferentiation mitochondrion organization myoblast differentiation response toprotozoan amino acidtransport leukocyte migration cytokine production t celldifferentiation protein secretion response tovirus angiogenesis Scrt1 Tcf25 Dpf1 Sap30 Ing2 Zfp654 Sp9 Zfp263 Mxi1 Hes6 Zfp395 Rlf Ppp1r13l Snapc1 C030039L03Rik Hif1a Arrb1 Glis3 Rcor2 Hif3a Fbxo21 Dnajc21 Rest Sirt6 Foxj1 Kdm5b Ankzf1 Sos2 Plscr1 Jdp2 Rbbp8 Etv4 Msh5 Mafg Tsc22d3 Nupr1 Ddit3 Cebpg Zfp790 Atf5 Cebpb Atf3 Trim21 Irf9 Irf2 Tbx21 Stat2 Stat1 Zbp1 Irf1 aGOslimPos Ikzf3 Oasl1 Irf7 Trim30a Dhx58 Txk Rorc Rora Nr1d2 Setdb2 Vdr Vax2 Nr1d1 Foxs1 Eno1 Thap3 Nfkbil1 Edf1 Srebf1 Lbr Tead1 Zfp608 Pcx Ift57 Ssbp4 Stat3 Mxd1 Pml Ssh2 Chd7 Maf Cic Bcl3 Prkdc Mbd5 Ppfibp2 Foxp2 Tal2 Mlf1 Bcl6b Zfp827 Ikzf2 Phtf2 Bmyc Plagl2 Nfkb2 Nfkb1 Tox Nrip1 Utf1 Klf3 Plagl1 Nfkbib Spib Nfkbie Akna Rbpj Ncoa3 Id1 Tnp2 Gata3 Gata1 Pparg Id2 Epas1 Zfp280b Commons Pathway Erg GO MSigDB KEGG Hhex WikiPathways SetGene Databases Batf Aff3 Zfp266 gene modules other (hypergeometric TF, from Figure Trim24 Zbtb5 Foxo3 Aebp2 XPodNet -protein-proteininteractionsinthepodocyteexpandedbySTRING Ppp1r10 Dffb Trp53 Enrichment
    [Show full text]
  • STEROID RECEPTORS in HEALTH and DISEASE SERONO SYMPOSIA, USA Series Editor: James Posillico
    STEROID RECEPTORS IN HEALTH AND DISEASE SERONO SYMPOSIA, USA Series Editor: James Posillico ACROMEGALY: A Century of Scientific and Clinical Progress Edited by Richard J. Robbins and Shlomo Melmed BASIC AND CLINICAL ASPECTS OF GROWTH HORMONE Edited by Barry B. Bercu THE PRIMATE OVARY Edited by Richard L. Stouffer SOMATOSTATIN: Basic and Clinical Status Edited by Seymour Reichlin STEROID RECEPTORS IN HEALTH AND DISEASE Edited by Virinder K. Moudgil A Continuation Order Plan is available for this series. A continuation order will bring delivery of each new volume immediately upon publication. Volumes are billed only upon actual shipment. For further infor­ mation please contact the publisher. STEROID RECEPTORS IN HEALTH AND DISEASE Edited by Virinder K. Moudgil Oakland University Rochester, Michigan PLENUM PRESS • NEW YORK AND LONDON library of Congress Cataloging in Publication Data Meadow Brook Conference on Steroid Receptors in Health and Disease (tst(tst:: t987 : Rochester, Mich .) Ste roid receptors in health and disease / edited by Virinder K. Moudgil. p. cm . "Proc" Proceedingseedings of the Meadow Brook Conference on Steroid Receptors in Health and Disease, spon­ sored by Serono Symposia. USA and Oakland University. held September 20-23, 19871987.. in Rochester. Michigan ··-T.p. verso. Includes bibliographies and indexes. IS8N 978-1-4684-5543-4 IS8N 978-1-4684-5541-0 (eBook) 00110.1007/978-1-4684-5541-0 I . Steroid hormones-Receptors-Congresses. 1. Moudgil, V.V. K.K. (Virinder K.), 1945- . 11. Sereno Symposia, USA. III.Ill. Oakland Universityly. IV. Title. [oNlM[oNlM:: tt. Receptors , Sleroid-congresses. W3 ME3781st 1987s / WK 150 M482 1987sJ1987sj oP571.7.M42 1987 512 '.
    [Show full text]