Integrative Genomic Analysis of Hnrnp L Splicing Regulation

Total Page:16

File Type:pdf, Size:1020Kb

Integrative Genomic Analysis of Hnrnp L Splicing Regulation University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2015 Terrae Incognitae: Integrative Genomic Analysis of Hnrnp L Splicing Regulation Brian Sebastian Cole University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Allergy and Immunology Commons, Biochemistry Commons, Bioinformatics Commons, Immunology and Infectious Disease Commons, and the Medical Immunology Commons Recommended Citation Cole, Brian Sebastian, "Terrae Incognitae: Integrative Genomic Analysis of Hnrnp L Splicing Regulation" (2015). Publicly Accessible Penn Dissertations. 1664. https://repository.upenn.edu/edissertations/1664 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/1664 For more information, please contact [email protected]. Terrae Incognitae: Integrative Genomic Analysis of Hnrnp L Splicing Regulation Abstract Alternative splicing is a critical component of human gene control that generates functional diversity from a limited genome. Defects in alternative splicing are associated with disease in humans. Alternative splicing is regulated developmentally and physiologically by the combinatorial actions of cis- and trans- acting factors, including RNA binding proteins that regulate splicing through sequence-specific interactions with pre-mRNAs. In T cells, the splicing regulator hnRNP L is an essential factor that regulates alternative splicing of physiologically important mRNAs, however the broader physical and functional impact of hnRNP L remains unknown. In this dissertation, I present analysis of hnRNP L-RNA interactions with CLIP-seq, which identifies transcriptome-wide binding sites and uncovers novel functional targets. I then use functional genomics studies to define pre-mRNA processing alterations induced by hnRNP L depletion, chief among which is cassette-type alternative splicing. Finally, I use integrative genomic analysis to identify a direct role for hnRNP L in repression of exon inclusion and an indirect role for enhancing exon inclusion that supports a novel regulatory interplay between hnRNP L and chromatin. In two appendices, I present CLIP-seq studies of two additional RNA binding proteins: the splicing factor CELF2 and the RNA helicase DDX17. In conclusion, I provide comparisons of these three CLIP-seq studies, providing high-level insights into the capabilities and limitations of CLIP-seq. In sum, this dissertation expands our knowledge of hnRNP L splicing control in the context of broader studies of RNA binding proteins in multiple cell types and conditions. Degree Type Dissertation Degree Name Doctor of Philosophy (PhD) Graduate Group Cell & Molecular Biology First Advisor Kristen W. Lynch Keywords Alternative splicing, CLIP-seq, hnRNP L, integrative genomics, RNA-seq, T cells Subject Categories Allergy and Immunology | Biochemistry | Bioinformatics | Immunology and Infectious Disease | Medical Immunology This dissertation is available at ScholarlyCommons: https://repository.upenn.edu/edissertations/1664 TERRAE INCOGNITAE: INTEGRATIVE GENOMIC ANALYSIS OF hnRNP L SPLICING REGULATION Brian Sebastian Cole A DISSERTATION in Cell and Molecular Biology Presented to the Faculties of the University of Pennsylvania in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy 2015 Supervisor of Dissertation ______________ Kristen W. Lynch, Ph.D. Professor of Biochemistry and Biophysics Graduate Group Chairperson _________________ Daniel S. Kessler, Ph.D., Associate Professor of Cell and Developmental Biology Dissertation Committee Russ P. Carstens, M.D., Associate Professor of Medicine Brian D. Gregory, Ph.D., Assistant Professor of Biology Yoseph Barash, Ph.D., Assistant Professor Genetics Stephen A. Liebhaber, M.D., Professor of Genetics and Medicine TERRAE INCOGNITAE: INTEGRATIVE GENOMIC ANALYSIS OF hnRNP L SPLICING REGULATION COPYRIGHT © 2015 Brian Sebastian Cole Permission is granted to copy, distribute, and/or modify this document under the terms of the GNU Free Documentation License, Version 1.3 or any later version published by the Free Software Foundation; with the Invariant Sections being DEDICATION and ACKNOWLEDGEMENTS, with the Front-Cover Texts being TITLE and COPYRIGHT, and with no Back-Cover Texts. To view a copy of this license, visit: https://www.gnu.org/licenses/fdl.html DEDICATION I dedicate this dissertation to my thesis advisor, Kristen Lynch. Her wisdom and strength of character will be with me always. iii ACKNOWLEDGMENT I acknowledge the many who have supported me in my research, both technically and personally. I’d like to thank the legions of open source programmers who have worked for Freedom. Without open source programs, I’d never have been able to take the journey I’ve taken over the past five years. I’d like to thank my family and friends for all their support over the past five years: my mom, my dad, my brother, Leksa, Mike, Ian, Roland, Sheila and Dan, Matt, the Venez Voir crew, Kyle and Andy and everybody at PBG, Rohit and Christina and everybody at WHIT, Craig and Charles over at Penn Health, Jordan and Bren and Luigi and the Data Incubator crew from New York City, Matt and the Beer Club, Mr. Nap the mathiest lawyer who ever drummed sevens and fives, and everybody else who has kept me afloat. iv ABSTRACT TERRAE INCOGNITAE: INTEGRATIVE GENOMIC ANALYSIS OF hnRNP L SPLICING REGULATION Brian Sebastian Cole Kristen W. Lynch, Ph.D. Alternative splicing is a critical component of human gene control that generates functional diversity from a limited genome. Defects in alternative splicing are associated with disease in humans. Alternative splicing is regulated developmentally and physiologically by the combinatorial actions of cis- and trans-acting factors, including RNA binding proteins that regulate splicing through sequence-specific interactions with pre-mRNAs. In T cells, the splicing regulator hnRNP L is an essential factor that regulates alternative splicing of physiologically important mRNAs, however the broader physical and functional impact of hnRNP L remains unknown. In this dissertation, I present analysis of hnRNP L-RNA interactions with CLIP-seq, which identifies transcriptome-wide binding sites and uncovers novel functional targets. I then use functional genomics studies to define pre-mRNA processing alterations induced by hnRNP L depletion, chief among which is cassette-type alternative splicing. Finally, I use integrative genomic analysis to identify a direct role for hnRNP L in repression of exon inclusion and an indirect role for enhancing exon inclusion that supports a novel regulatory interplay between hnRNP L and chromatin. In two appendices, I present CLIP-seq studies of two additional RNA binding proteins: the splicing factor CELF2 and the RNA helicase DDX17. In conclusion, I provide comparisons of these three CLIP-seq studies, providing high-level insights into the capabilities and limitations of CLIP-seq. In v sum, this dissertation expands our knowledge of hnRNP L splicing control in the context of broader studies of RNA binding proteins in multiple cell types and conditions. vi TABLE OF CONTENTS ACKNOWLEDGMENT .............................................................................................................IV ABSTRACT.................................................................................................................................. V LIST OF TABLES ....................................................................................................................... X LIST OF FIGURES.....................................................................................................................XI INTRODUCTION ....................................................................................................................... 1 The spliceosome catalyzes splicing through serial assembly and rearrangements ............. 1 Regulation of splicing is achieved by RNA binding proteins................................................... 2 hnRNP L is an essential regulator of alternative splicing in T cells ........................................ 6 Technologies for transcriptome-wide investigation of RNA binding proteins ..................... 10 Functional studies of splicing factors with next-generation sequencing reveal splicing regulatory activity....................................................................................................................... 11 MATERIALS AND METHODS...............................................................................................14 CLIP-seq read processing and alignment ................................................................................ 14 CLIP-seq binding site definition: peak calling ......................................................................... 17 CLIP-seq motif enrichment analysis ......................................................................................... 21 CLIP-seq peak overlap comparison.......................................................................................... 23 Gene ontology analysis.............................................................................................................. 24 Splicing analysis from RASL-seq data ..................................................................................... 24 Splicing analysis from mRNA-seq data.................................................................................... 25 Stringent union of
Recommended publications
  • Topological Scoring of Protein Interaction Networks
    bioRxiv preprint doi: https://doi.org/10.1101/438408; this version posted October 8, 2018. 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. Topological Scoring of Protein Interaction Networks Mihaela E. Sardiu1, Joshua M. Gilmore1,2, Brad D. Groppe1,3, Arnob Dutta1,4, Laurence Florens1, and Michael P. Washburn1,5‡ 1Stowers Institute for Medical Research, Kansas City, MO 64110 U.S.A. 2Current Address: Boehringer Ingelheim Vetmedica, St. Joseph, MO 64506 U.S.A. 3Current Address: Thermo Fisher Scientific, Waltham, MA 02451, U.S.A. 4Current Address: Department of Cell and Molecular Biology, University of Rhode Island, 287 CBLS, 120 Flagg Road, Kingston, RI 02881. 5 Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, Kansas 66160, USA ‡To whom correspondence should be addressed: Michael Washburn, Ph.D. Stowers Institute for Medical Research 1000 E. 50th St. Kansas City, MO 64110 Phone: 816-926-4457 E-mail: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/438408; this version posted October 8, 2018. 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 It remains a significant challenge to define individual protein associations within networks where an individual protein can directly interact with other proteins and/or be part of large complexes, which contain functional modules. Here we demonstrate the topological scoring (TopS) algorithm for the analysis of quantitative proteomic analyses of affinity purifications.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Targets of Hnrnp L in Human T Cells Profiling Reveals Physical And
    Transcriptome-Wide RNA Interaction Profiling Reveals Physical and Functional Targets of hnRNP L in Human T Cells Downloaded from Ganesh Shankarling, Brian S. Cole, Michael J. Mallory and Kristen W. Lynch Mol. Cell. Biol. 2014, 34(1):71. DOI: 10.1128/MCB.00740-13. Published Ahead of Print 28 October 2013. http://mcb.asm.org/ Updated information and services can be found at: http://mcb.asm.org/content/34/1/71 These include: SUPPLEMENTAL MATERIAL Supplemental material on December 12, 2013 by UNIVERSITY OF PENNSYLVANIA LIBRARY REFERENCES This article cites 58 articles, 30 of which can be accessed free at: http://mcb.asm.org/content/34/1/71#ref-list-1 CONTENT ALERTS Receive: RSS Feeds, eTOCs, free email alerts (when new articles cite this article), more» Information about commercial reprint orders: http://journals.asm.org/site/misc/reprints.xhtml To subscribe to to another ASM Journal go to: http://journals.asm.org/site/subscriptions/ Transcriptome-Wide RNA Interaction Profiling Reveals Physical and Functional Targets of hnRNP L in Human T Cells Downloaded from Ganesh Shankarling, Brian S. Cole, Michael J. Mallory, Kristen W. Lynch ‹Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA The RNA processing factor hnRNP L is required for T cell development and function. However, the spectrum of direct targets of hnRNP L activity in T cells has yet to be defined. In this study, we used cross-linking and immunoprecipitation followed by high- throughput sequencing (CLIP-seq) to identify the RNA binding sites of hnRNP L within the transcriptomes of human CD4؉ and cultured Jurkat T cells.
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • Nuclear PTEN Safeguards Pre-Mrna Splicing to Link Golgi Apparatus for Its Tumor Suppressive Role
    ARTICLE DOI: 10.1038/s41467-018-04760-1 OPEN Nuclear PTEN safeguards pre-mRNA splicing to link Golgi apparatus for its tumor suppressive role Shao-Ming Shen1, Yan Ji2, Cheng Zhang1, Shuang-Shu Dong2, Shuo Yang1, Zhong Xiong1, Meng-Kai Ge1, Yun Yu1, Li Xia1, Meng Guo1, Jin-Ke Cheng3, Jun-Ling Liu1,3, Jian-Xiu Yu1,3 & Guo-Qiang Chen1 Dysregulation of pre-mRNA alternative splicing (AS) is closely associated with cancers. However, the relationships between the AS and classic oncogenes/tumor suppressors are 1234567890():,; largely unknown. Here we show that the deletion of tumor suppressor PTEN alters pre-mRNA splicing in a phosphatase-independent manner, and identify 262 PTEN-regulated AS events in 293T cells by RNA sequencing, which are associated with significant worse outcome of cancer patients. Based on these findings, we report that nuclear PTEN interacts with the splicing machinery, spliceosome, to regulate its assembly and pre-mRNA splicing. We also identify a new exon 2b in GOLGA2 transcript and the exon exclusion contributes to PTEN knockdown-induced tumorigenesis by promoting dramatic Golgi extension and secretion, and PTEN depletion significantly sensitizes cancer cells to secretion inhibitors brefeldin A and golgicide A. Our results suggest that Golgi secretion inhibitors alone or in combination with PI3K/Akt kinase inhibitors may be therapeutically useful for PTEN-deficient cancers. 1 Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China. 2 Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences and SJTU-SM, Shanghai 200025, China.
    [Show full text]
  • Extracellular Matrix Protein-1 Secretory Isoform Promotes Ovarian Cancer Through Increasing Alternative Mrna Splicing and Stemness
    ARTICLE https://doi.org/10.1038/s41467-021-24315-1 OPEN Extracellular matrix protein-1 secretory isoform promotes ovarian cancer through increasing alternative mRNA splicing and stemness Huijing Yin1,2,8, Jingshu Wang3,8, Hui Li3,8, Yinjue Yu3,8, Xiaoling Wang4, Lili Lu1,2, Cuiting Lv3, Bin Chang2,5, ✉ ✉ Wei Jin6, Wenwen Guo2,7, Chunxia Ren 4 & Gong Yang 1,2,3 1234567890():,; Extracellular matrix protein-1 (ECM1) promotes tumorigenesis in multiple organs but the mechanisms associated to ECM1 isoform subtypes have yet to be clarified. We report in this study that the secretory ECM1a isoform induces tumorigenesis through the GPR motif binding to integrin αXβ2 and the activation of AKT/FAK/Rho/cytoskeleton signaling. The ATP binding cassette subfamily G member 1 (ABCG1) transduces the ECM1a-integrin αXβ2 interactive signaling to facilitate the phosphorylation of AKT/FAK/Rho/cytoskeletal molecules and to confer cancer cell cisplatin resistance through up-regulation of the CD326- mediated cell stemness. On the contrary, the non-secretory ECM1b isoform binds myosin and blocks its phosphorylation, impairing cytoskeleton-mediated signaling and tumorigenesis. Moreover, ECM1a induces the expression of the heterogeneous nuclear ribonucleoprotein L like (hnRNPLL) protein to favor the alternative mRNA splicing generating ECM1a. ECM1a, αXβ2, ABCG1 and hnRNPLL higher expression associates with poor survival, while ECM1b higher expression associates with good survival. These results highlight ECM1a, integrin αXβ2, hnRNPLL and ABCG1 as potential targets for treating cancers associated with ECM1- activated signaling. 1 Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China. 2 Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, China.
    [Show full text]
  • MEIS2 Promotes Cell Migration and Invasion in Colorectal Cancer
    ONCOLOGY REPORTS 42: 213-223, 2019 MEIS2 promotes cell migration and invasion in colorectal cancer ZIANG WAN1*, RUI CHAI1*, HANG YUAN1*, BINGCHEN CHEN1, QUANJIN DONG1, BOAN ZHENG1, XIAOZHOU MOU2, WENSHENG PAN3, YIFENG TU4, QING YANG5, SHILIANG TU1 and XINYE HU1 1Department of Colorectal Surgery, 2Clinical Research Institute and 3Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014; 4Department of Pathology, College of Basic Medical Sciences, Shenyang Medical College, Shenyang, Liaoning 110034; 5Department of Academy of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China Received October 9, 2018; Accepted March 18, 2019 DOI: 10.3892/or.2019.7161 Abstract. Colorectal cancer (CRC) is one of the most common 5-year survival rate of metastatic CRC is as low as ~10%. In types of malignancy worldwide. Distant metastasis is a key the past few decades, several regulators of CRC metastasis cause of CRC‑associated mortality. MEIS2 has been identified have been identified, including HNRNPLL and PGE2 (4,5). to be dysregulated in several types of human cancer. However, HNRNPLL has been revealed to modulate alternative splicing the mechanisms underlying the regulatory role of MEIS2 in of CD44 during the epithelial-mesenchymal transition (EMT), CRC metastasis remain largely unknown. For the first time, which leads to suppression of CRC metastasis (4). PGE2 the present study demonstrated that MEIS2 serves a role as a induced an expansion of CRC stem cells to promote liver promoter of metastasis in CRC. In vivo and in vitro experiments metastases in mice by activating NF-κB (5).
    [Show full text]
  • A Genome-Wide Resource and Visualization Tool to Design CRISPR/Cas9 Screens for Editing Protein-RNA Interaction Sites in the Human Genome
    bioRxiv preprint doi: https://doi.org/10.1101/654640; this version posted September 3, 2019. 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. SliceIt: A genome-wide resource and visualization tool to design CRISPR/Cas9 screens for editing protein-RNA interaction sites in the human genome Sasank Vemuri1Ψ, Rajneesh Srivastava1Ψ, Quoseena Mir1, Seyedsasan Hashemikhabir1, X. Charlie Dong2, 1, 3, 4* Sarath Chandra Janga 1Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University, 719 Indiana Ave Ste 319, Walker Plaza Building, Indianapolis, Indiana 46202 2Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, 635 Barnhill Drive, Indianapolis, Indiana, 46202 3Department of Medical and Molecular Genetics, Indiana University School of Medicine, Medical Research and Library Building, 975 West Walnut Street, Indianapolis, Indiana, 46202 4Centre for Computational Biology and Bioinformatics, Indiana University School of Medicine, 5021 Health Information and Translational Sciences (HITS), 410 West 10th Street, Indianapolis, Indiana, 46202 ΨBoth the authors contributed equally to this study and should be considered as joint first authors Keywords: RNA binding proteins, In silico library, CRISPR/Cas9, Guide RNA, Pooled CRISPR screens, Protein-RNA interactions, Post-transcriptional control *Correspondence should be addressed to: Sarath Chandra Janga ([email protected]) 719 Indiana Avenue Ste 319, Walker Plaza Building Indianapolis, Indiana – 46202 Tel: +1-317-278-4147, Fax: +1-317-278-9201 bioRxiv preprint doi: https://doi.org/10.1101/654640; this version posted September 3, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.
    [Show full text]
  • Estrogen Regulation and Physiopathologic Significance of Alternative Promoters in Breast Cancer
    Published OnlineFirst April 20, 2010; DOI: 10.1158/0008-5472.CAN-09-3988 Published OnlineFirst on April 20, 2010 as 10.1158/0008-5472.CAN-09-3988 Tumor and Stem Cell Biology Cancer Research Estrogen Regulation and Physiopathologic Significance of Alternative Promoters in Breast Cancer Martin Dutertre1, Lise Gratadou1, Etienne Dardenne1, Sophie Germann1, Samaan Samaan1, Rosette Lidereau2, Keltouma Driouch2, Pierre de la Grange3, and Didier Auboeuf1 Abstract Alternative promoters (AP) occur in >30% protein-coding genes and contribute to proteome diversity. How- ever, large-scale analyses of AP regulation are lacking, and little is known about their potential physiopatho- logic significance. To better understand the transcriptomic effect of estrogens, which play a major role in breast cancer, we analyzed gene and AP regulation by estradiol in MCF7 cells using pan-genomic exon arrays. We thereby identified novel estrogen-regulated genes (ERG) and determined the regulation of AP-encoded transcripts in 150 regulated genes. In <30% cases, APs were regulated in a similar manner by estradiol, whereas in >70% cases, they were regulated differentially. The patterns of AP regulation correlated with the patterns of estrogen receptor α (ERα) and CCCTC-binding factor (CTCF) binding sites at regulated gene loci. Interest- ingly, among genes with differentially regulated (DR) APs, we identified cases where estradiol regulated APs in an opposite manner, sometimes without affecting global gene expression levels. This promoter switch was mediated by the DDX5/DDX17 family of ERα coregulators. Finally, genes with DR promoters were preferen- tially involved in specific processes (e.g., cell structure and motility, and cell cycle). We show, in particular, that isoforms encoded by the NET1 gene APs, which are inversely regulated by estradiol, play distinct roles in cell adhesion and cell cycle regulation and that their expression is differentially associated with prognosis in ER+ breast cancer.
    [Show full text]
  • DDX17 Specifically, and Independently of DDX5, Controls Use of the HIV A4/5 Splice Acceptor Cluster and Is Essential for Efficient Replication of HIV
    Article DDX17 Specifically, and Independently of DDX5, Controls Use of the HIV A4/5 Splice Acceptor Cluster and Is Essential for Efficient Replication of HIV Nyaradzai Sithole 1, Claire A. Williams 1,†, Aisling M. Vaughan 1,‡, Julia C. Kenyon 1,2 and Andrew M.L. Lever 1,3 1 - Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK 2 - Department of Microbiology and Immunology, National University of Singapore, Singapore 117545 3 - Department of Medicine, National University of Singapore, Singapore 119228 Correspondence to Andrew M.L. Lever: Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK. [email protected] https://doi.org/10.1016/j.jmb.2018.06.052 Edited by Eric O. Freed Abstract HIV splicing involves five splice donor and eight splice acceptor sequences which, together with cryptic splice sites, generate over 100 mRNA species. Ninety percent of both partially spliced and fully spliced transcripts utilize the intrinsically weak A4/A5 3′ splice site cluster. We show that DDX17, but not its close paralog DDX5, specifically controls the usage of this splice acceptor group. In its absence, production of the viral envelope protein and other regulatory and accessory proteins is grossly reduced, while Vif, which uses the A1 splice acceptor, is unaffected. This is associated with a profound decrease in viral export from the cell. Loss of Vpu expression causing upregulation of cellular Tetherin compounds the phenotype. DDX17 utilizes distinct RNA binding motifs for its role in efficient HIV replication, and we identify RNA binding motifs essential for its role, while the Walker A, Walker B (DEAD), Q motif and the glycine doublet motif are all dispensable.
    [Show full text]
  • Identification of Protein Features Encoded by Alternative Exons Using Exon Ontology
    Downloaded from genome.cshlp.org on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press Resource Identification of protein features encoded by alternative exons using Exon Ontology Léon-Charles Tranchevent,1 Fabien Aubé,1 Louis Dulaurier,1 Clara Benoit-Pilven,1 Amandine Rey,1 Arnaud Poret,1 Emilie Chautard,2 Hussein Mortada,1 François-Olivier Desmet,1 Fatima Zahra Chakrama,1 Maira Alejandra Moreno-Garcia,1 Evelyne Goillot,3 Stéphane Janczarski,1 Franck Mortreux,1 Cyril F. Bourgeois,1,4 and Didier Auboeuf1,4 1Université Lyon 1, ENS de Lyon, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France; 2Laboratoire de Biométrie et Biologie Évolutive, Université Lyon 1, UMR CNRS 5558, INRIA Erable, Villeurbanne, F-69622, France; 3Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Lyon 1, Lyon, F-69007 France Transcriptomic genome-wide analyses demonstrate massive variation of alternative splicing in many physiological and pathological situations. One major challenge is now to establish the biological contribution of alternative splicing var- iation in physiological- or pathological-associated cellular phenotypes. Toward this end, we developed a computational approach, named Exon Ontology, based on terms corresponding to well-characterized protein features organized in an ontology tree. Exon Ontology is conceptually similar to Gene Ontology-based approaches but focuses on exon-encod- ed protein features instead of gene level functional annotations. Exon Ontology describes the protein features encoded by a selected list of exons and looks for potential Exon Ontology term enrichment. By applying this strategy to exons that are differentially spliced between epithelial and mesenchymal cells and after extensive experimental validation, we demonstrate that Exon Ontology provides support to discover specific protein features regulated by alternative splic- ing.
    [Show full text]
  • The DEAD-Box Protein P72 Regulates ERΑ-&Sol;Oestrogen
    Oncogene (2009) 28, 4053–4064 & 2009 Macmillan Publishers Limited All rights reserved 0950-9232/09 $32.00 www.nature.com/onc ORIGINAL ARTICLE The DEAD-box protein p72 regulates ERa-/oestrogen-dependent transcription and cell growth, and is associated with improved survival in ERa-positive breast cancer NC Wortham1,6, E Ahamed2,6, SM Nicol1, RS Thomas2, M Periyasamy2, J Jiang2,3, AM Ochocka2, S Shousha3, L Huson4, SE Bray5, RC Coombes2, S Ali2 and FV Fuller-Pace1 1Centre for Oncology and Molecular Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK; 2Department of Oncology, Imperial College London, Hammersmith Hospital Campus, London, UK; 3Department of Histopathology, Imperial College London, Charing Cross Hospital, London, UK; 4Statistical Advisory Service, Imperial College London, South Kensington Campus, London, UK and 5Tayside Tissue Bank, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK The DEAD-box RNA helicases p68 (DDX5) and p72 Introduction (DDX17) have been shown to act as transcriptional co- activators for a diverse range of transcription factors, The DEAD-box subfamily of RNA helicases, originally including oestrogen receptor-a (ERa). Here, we show that, so named on the basis of the presence of a conserved although both proteins interact with and co-activate ERa motif having the sequence Asp-Glu-Ala-Asp, have been in reporter gene assays, small interfering RNA-mediated implicated in cellular processes involving the regulation knockdown of p72, but not p68, results in a significant of RNA structure, including pre-mRNA processing, inhibition of oestrogen-dependent transcription of endo- RNA export, RNA degradation, ribosome assembly, genous ERa-responsive genes and oestrogen-dependent translation and miRNA maturation (Linder et al., 1989; growth of MCF-7 and ZR75-1 breast cancer cells.
    [Show full text]