Rna Pol Ii Cleavage Activity Is a Major Effector of Transcription

Total Page:16

File Type:pdf, Size:1020Kb

Rna Pol Ii Cleavage Activity Is a Major Effector of Transcription RNA POL II CLEAVAGE ACTIVITY IS A MAJOR EFFECTOR OF TRANSCRIPTION DYNAMICS by RYAN MICHAEL SHERIDAN B.S., University of Missouri-St. Louis, 2012 A thesis submitted to the Faculty of the Graduate School of the University of Colorado in partial fulfillment of the requirements for the degree of Doctor of Philosophy Molecular Biology Program 2019 This thesis for the Doctor of Philosophy degree by Ryan Michael Sheridan has been approved for the Molecular Biology Program by Sandy Martin, Chair Richard Davis Jay Hesselberth Thomas Blumenthal David Barton David Bentley, Advisor Date: May 17, 2019 ii Sheridan, Ryan Michael (Ph.D., Molecular Biology) RNA Pol II Cleavage Activity is a Major Effector of Transcription Dynamics Thesis directed by Professor David L. Bentley ABSTRACT After transcription initiation, RNA polymerase II (pol II) undergoes several steps that can influence gene expression, including promoter proximal pausing, transcription elongation, and termination. Promoter proximal pausing occurs after pol II has moved 50- 100 bp into the gene body and is induced by negative elongation factors that interact with the polymerase. After release from the 5’ pause, the rate of pol II elongation increases as the polymerase moves further from the TSS. Upon reaching the poly(A) site, transcription termination occurs at a position influenced by a kinetic competition between pol II and termination factor Xrn2. The mechanisms that influence the release of 5’ paused pol II and subsequent elongation rates within gene bodies and downstream of poly(A) sites are poorly understood, but one potential rate-limiting event is pol II backtracking. Pol II backtracking can occur when the polymerase mis-incorporates a base or encounters a roadblock that causes the polymerase to move backwards, dislodging the nascent RNA 3’ end from the active site. To rescue backtracked pol II, the transcription factor TFIIS stimulates the RNA endonuclease activity of the polymerase, which cleaves the nascent RNA to generate a new correctly positioned 3’ end. To investigate the importance of pol II cleavage activity in the transcription cycle, I used a dominant negative TFIIS mutant (TFIISDN) that inhibits this cleavage activity. Using this mutant, I mapped individual pol II pause and backtrack sites and find that these events occur throughout iii transcription but are especially enriched at the 5’ and 3’ ends of genes. I find that inhibition of pol II cleavage activity impairs the release of 5’ paused polymerases and causes transcription to terminate at a more proximal position downstream of the poly(A) site. In addition, expression of TFIISDN results in a ~50% reduction in the rate of elongation and delays the upregulation of hypoxia and heat shock responsive transcripts. These results demonstrate that pol II backtracking is widespread, and the intrinsic RNA cleavage activity of the polymerase influences each post-initiation step of the transcription cycle. The ability to deplete endogenous human proteins is valuable for studying cellular processes. Traditional protein depletion methods such as shRNA-mediated knockdown suffer from off-target effects, incomplete knockdown, and slow kinetics. In addition, if the protein of interest is required for cell viability, a conditional knockdown method must be applied. Protein destabilization domains known as degrons present an appealing strategy for achieving conditional depletion of endogenous factors in a rapid and complete manner. I developed a PCR-based method for tagging endogenous human proteins with an eDHFR degron tag. The stability of eDHFR-tagged proteins can be controlled by modulating the concentration of trimethoprim in the culture media. This approach allows for rapid depletion of eDHFR-tagged proteins that is both reversible and complete. The form and content of this abstract are approved. I recommend its publication. Approved: David L. Bentley iv DEDICATION This thesis is dedicated to my family and friends. To my parents who have always supported my endeavors and pushed me to live a happy, healthy, and balanced life. To my sister who is always willing to help me escape the city and journey into the mountains. To my fiancée, Alexis, who has been with me through this entire strange trip and is never afraid to tell a bad joke to lighten my day. To my loving grandparents who are so good at spoiling me with delicious cooking and good conversation. To my aunts, uncles, and cousins who are always willing to share their homes for our summer retreats. To my friends who have provided me with so much laughter and absurdity, and a countless number of misadventures. These people have supported me so much through this journey and I am forever grateful that they have been a part of my life. v ACKNOWLEDGEMENTS I would like to thank my thesis advisor, David Bentley, who has provided me with so much advice and guidance. Your leadership has fostered such a positive and supportive work environment. I am so proud to have been a part of your lab and I can honestly say that these have been some of the most exciting and rewarding years of my life. I also want to thank members of the Bentley lab including Nova, Michael, Tassa, Ben, and Hyunmin. Nova, not only did you generate the TFIIS cell lines used for my thesis, but you were also responsible for training me in so many techniques. I want to thank Michael and Tassa for their helpful suggestions about the project, and Ben and Hyunmin for their bioinformatics help when I first joined David’s lab. I would also like to thank my thesis committee for their insightful suggestions and for taking the time to keep me on track. During my time as a graduate student I took several courses and workshops that were especially helpful to my project. This included Jay Hesselberth’s Genome Informatics Workshop and a GRO-seq workshop run by Mary Allen and Robin Dowell. I would also like to thank my undergraduate advisor, Teresa Thiel and her lab manager Brenda Pratte. It’s hard for me to imagine that I would be where I am today if not for my experience in the Thiel lab. I would like to thank the Biochemistry and Molecular Genetics Department for bringing together such a great group of scientists and providing an incredibly collaborative work environment. I would like to thank the Molecular Biology Program for taking me in and funding my first three years of graduate school. I would also like to thank the Bolie and Hirs families for providing funding for me to attend so many great conferences. I would like to thank the RNA Bioscience Initiative for all the work they vi have done to advance RNA research and for funding my project for the last two years. Lastly, I would like to thank Jingshi Shen for providing me with the HAP1 cells used for chapter V of my thesis. vii TABLE OF CONTENTS CHAPTER I. INTRODUCTION .................................................................................................. 1 1. Transcription by RNA polymerase II ...................................................................... 1 2. Mechanism of RNA polymerase backtracking ...................................................... 2 3. Regulation of promoter proximal pol II pausing .................................................. 10 4. Pol II pausing and backtracking during transcription elongation ......................... 15 5. Control of transcription termination ..................................................................... 19 6. The transcriptional response to hypoxia ............................................................. 22 7. The transcriptional heat shock response ............................................................ 24 8. Methods for depleting human proteins ............................................................... 25 II. MATERIALS AND METHODS ............................................................................ 30 1. Human cell lines ................................................................................................. 30 2. Antibodies ........................................................................................................... 30 3. Immunoblotting ................................................................................................... 31 4. Flow cytometry ................................................................................................... 31 5. ChIP-seq ............................................................................................................ 31 5A. Immunoprecipitation and library preparation ........................................................ 31 5B. Quantification of ChIP-seq signals ....................................................................... 32 6. Bru-seq ............................................................................................................... 33 viii 7. Calculation of elongation rates ........................................................................... 35 8. mNET-seq .......................................................................................................... 36 8A. Immunoprecipitation and library preparation ........................................................ 36 8B. Identification of pol II pause sites ......................................................................... 37 8C. Cross-correlation analysis .................................................................................... 38 9. GRO-seq ............................................................................................................ 38 10. Calculation of
Recommended publications
  • BRCA1-Dependent Transcriptional Regulation: Implication in Tissue-Specific Tumor Suppression
    cancers Review BRCA1-Dependent Transcriptional Regulation: Implication in Tissue-Specific Tumor Suppression Xiaowen Zhang * and Rong Li * Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA * Correspondence: [email protected] (X.Z.); [email protected] (R.L.) Received: 14 October 2018; Accepted: 11 December 2018; Published: 14 December 2018 Abstract: Germ-line mutations in breast cancer susceptibility gene 1 (BRCA1) predominantly predispose women to breast and ovarian cancers. BRCA1 is best known for its functions in maintenance of genomic integrity including repairing DNA double-strand breaks through homologous recombination and suppressing DNA replication stress. However, whether these universally important BRCA1 functions in maintenance of genomic stability are sufficient to account for its tissue-specific tumor-suppressing function remains unclear. Accumulating evidence indicates that there are previously underappreciated roles of BRCA1 in transcriptional regulation and chromatin remodeling. In this review, we discuss the functional significance of interactions between BRCA1 and various transcription factors, its role in epigenetic regulation and chromatin dynamics, and BRCA1-dependent crosstalk between the machineries of transcription and genome integrity. Furthermore, we propose a model of how transcriptional regulation could contribute to tissue-dependent tumor-suppressing function of BRCA1. Keywords: BRCA1; transcriptional regulation; epigenetic regulation; chromatin organization 1. Introduction Approximately 0.2% to 0.3% of the general population in the United States carries germ-line mutations in the tumor suppressor gene BRCA1 (BRCA1mut/+)[1,2]. Unlike tumor suppressors such as p53 that are implicated in a broad spectrum of cancers, BRCA1 functions in a gender- and tissue-specific manner.
    [Show full text]
  • Cofactor of BRCA1: a New Genetic Marker for Common Malignant Liver Cancer
    RESEARCH HIGHLIGHTS doi: 10.18282/amor.v2.i4.157 Cofactor of BRCA1: A new genetic marker for common malignant liver cancer new study has identified a vital gene in the pathogenesis and progression of liver cancer hepatocellular carcinoma (HCC), according to a A team of researchers at The American University in Cairo, Egypt, in a paper published in this issue of AMOR. The study on human gene ‘cofactor of BRCA1’ (dubbed COBRA1) and its potential role as a reliable cancer predictor for HCC is especially important owing to the disease’s grim outlook. HCC is “ranked as the second most common cause of cancer-related deaths in the world in 2012,” the authors said. “Thus, it is considered as a highly aggressive cancer with poor prognosis,” they added. The behavior of COBRA1 in the development and pro- gression of several cancers has previously been studied and established. Youssef observed, “For example, cell lines and tissues isolated from late-stage metastatic breast cancer tumors showed low expression levels of the COBRA1 protein, which is known to display tumor suppressor activ- ity.” In contrast, “another study reported that COBRA1 was overexpressed in upper gastrointestinal carcinoma (UGC) tissue samples,” they added. Yet, COBRA1’s involvement in HCC tumor formation and growth has been subjected to minimal studies. “To date, the molecular mechanisms underlying HCC patho- genesis have not been fully identified, thus resulting in a lack of reliable prognostic markers for HCC,” said the re- According to data from the Surveillance Epidemiology searchers. and End Results (SEER) program, hepatocellular carcino- ma accounts for 90% of all liver cancers worldwide.
    [Show full text]
  • NELF-Mediated Stalling of Pol II Can Enhance Gene Expression by Blocking Promoter-Proximal Nucleosome Assembly
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press NELF-mediated stalling of Pol II can enhance gene expression by blocking promoter-proximal nucleosome assembly Daniel A. Gilchrist,1 Sergei Nechaev,1 Chanhyo Lee,2 Saikat Kumar B. Ghosh,2 Jennifer B. Collins,3 Leping Li,4 David S. Gilmour,2 and Karen Adelman1,3,5 1Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA; 2Department of Biochemistry and Molecular Biology, Center for Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania 16802, USA; 3Microarray Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA; 4Biostatistics Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA The Negative Elongation Factor (NELF) is a transcription regulatory complex that induces stalling of RNA polymerase II (Pol II) during early transcription elongation and represses expression of several genes studied to date, including Drosophila Hsp70, mammalian proto-oncogene junB, and HIV RNA. To determine the full spectrum of NELF target genes in Drosophila, we performed a microarray analysis of S2 cells depleted of NELF and discovered that NELF RNAi affects many rapidly inducible genes involved in cellular responses to stimuli. Surprisingly,
    [Show full text]
  • Alternative Splicing: Role in Cancer Development and Progression
    International Journal of Cell Biology Alternative Splicing: Role in Cancer Development and Progression Guest Editors: Claudia Ghigna, Michael Ladomery, and Claudio Sette Alternative Splicing: Role in Cancer Development and Progression International Journal of Cell Biology Alternative Splicing: Role in Cancer Development and Progression Guest Editors: Claudia Ghigna, Michael Ladomery, and Claudio Sette Copyright © 2013 Hindawi Publishing Corporation. All rights reserved. This is a special issue published in “International Journal of Celllogy.” Bio All articles are open access articles distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Editorial Board Paul N. Adler, USA Wiljan Hendriks, The Netherlands Liza Pon, USA Emad Alnemri, USA Paul J. Higgins, USA Jerome Rattner, Canada Avri Ben-Ze’ev, Israel Michael Hortsch, USA Maria Roubelakis, Greece Jeannette Chloe Bulinski, USA Pavel Hozak, Czech Republic Afshin Samali, Ireland Michael Bustin, USA Jeremy Hyams, France Michael Peter Sarras, USA John Cooper, USA Anton M. Jetten, USA Hirofumi Sawai, Japan Adrienne D. Cox, USA Edward M. Johnson, USA R. Seger, Israel J. R. Davie, Canada Daniel P. Kiehart, USA Barry D. Shur, USA Govindan Dayanithi, France Sharad Kumar, Australia Arnoud Sonnenberg, The Netherlands Arun M. Dharmarajan, Australia Paul Marks, USA Gary S. Stein, USA Dara Dunican, Ireland Seamus J. Martin, Ireland Tung Tien Sun, USA William Dunn, USA Manuela Martins-Green, USA Ming Tan, USA Victor Faundez, USA Takeshi Noda, Japan Guido Tarone, Italy Roland Foisner, Austria Helga Ogmundsd¨ ottir,´ Iceland Jean-Pierre Tassan, France Hans Hermann Gerdes, Norway Shoichiro Ono, USA Richard Tucker, USA Richard Gomer, USA Howard Beverley Osborne, France Andre Van Wijnen, USA Hinrich Gronemeyer, France Markus Paulmichl, Austria Gerhard Wiche, Austria Mehran Haidari, USA H.
    [Show full text]
  • Gene-Specific Genetic Complementation Between Brca1
    www.nature.com/scientificreports OPEN Gene-Specifc Genetic Complementation between Brca1 and Cobra1 During Mouse Received: 10 October 2017 Accepted: 29 January 2018 Mammary Gland Development Published: xx xx xxxx Huai-Chin Chiang1, Xiaowen Zhang1, Xiayan Zhao2, Chi Zhang 1, Jerry Chen1, Paula Garza1, Sabrina Smith1, Thomas Ludwig3, Richard J. Baer4, Rong Li1 & Yanfen Hu1 Germ-line mutations in breast cancer susceptibility gene, BRCA1, result in familial predisposition to breast and ovarian cancers. The BRCA1 protein has multiple functional domains that interact with a variety of proteins in multiple cellular processes. Understanding the biological consequences of BRCA1 interactions with its binding partners is important for elucidating its tissue-specifc tumor suppression function. The Cofactor of BRCA1 (COBRA1) is a BRCA1-binding protein that, as a component of negative elongation factor (NELF), regulates RNA polymerase II pausing during transcription elongation. We recently identifed a genetic interaction between mouse Brca1 and Cobra1 that antagonistically regulates mammary gland development. However, it remains unclear which of the myriad functions of Brca1 are required for its genetic interaction with Cobra1. Here, we show that, unlike deletion of Brca1 exon 11, separation-of-function mutations that abrogate either the E3 ligase activity of its RING domain or the phospho-recognition property of its BRCT domain are not sufcient to rescue the mammary developmental defects in Cobra1 knockout mice. Furthermore, deletion of mouse Palb2, another breast cancer susceptibility gene with functional similarities to BRCA1, does not rescue Cobra1 knockout-associated mammary defects. Thus, the Brca1/Cobra1 genetic interaction is both domain- and gene-specifc in the context of mammary gland development.
    [Show full text]
  • Regulation of Clustered Gene Expression by Cofactor of BRCA1 (COBRA1) in Breast Cancer Cells
    Oncogene (2007) 26, 2543–2553 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc ORIGINAL ARTICLE Regulation of clustered gene expression by cofactor of BRCA1 (COBRA1) in breast cancer cells SE Aiyar1, AL Blair1, DA Hopkinson, S Bekiranov and R Li Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA, USA Eucaryotic genes that are coordinately expressed tend to the gene organization patterns within a genome. In be clustered. Furthermore, gene clusters across chromo- contrast to the long-held presumption that genes are somal regions are often upregulated in various tumors. randomly distributed throughout the eucaryotic ge- However, relatively little is known about how gene nomes, there is compelling evidence for clustering of clusters are coordinately expressed in physiological or co-expressed genes in every eucaryotic genome exam- pathological conditions. Cofactor of BRCA1 (COBRA1), ined so far (Hurst et al., 2004). For example, 25% of a subunit of the human negative elongation factor, has genes in budding yeast that are transcribed in the same been shown to repress estrogen-stimulated transcription cell-cycle stage are adjacently located (Cho et al., 1998). of trefoil factor 1 (TFF1 or pS2)by stalling RNA The gene-clustering phenomenon is perhaps even more polymerase II. Here, we carried out a genome-wide study prominent in multicellular organisms. It is reported that to identify additional physiological target genes of 20% of genes in the entire fly genome are organized into COBRA1 in breast cancer cells. The study identified a clusters with similar expression patterns (Spellman and total of 134 genes that were either activated or repressed Rubin, 2002).
    [Show full text]
  • Cofactor of BRCA1: a Novel Transcription Factor Regulator in Upper Gastrointestinal Adenocarcinomas
    Research Article Cofactor of BRCA1: A Novel Transcription Factor Regulator in Upper Gastrointestinal Adenocarcinomas Patricia A. McChesney,1 Sarah E. Aiyar,2 Ok-Jae Lee,1,4,5 Alexander Zaika,1,5 Christopher Moskaluk,3 Rong Li,2 and Wa’el El-Rifai1,5 Departments of 1Internal Medicine, 2Biochemistry and Molecular Genetics, and 3Pathology, University of Virginia, Charlottesville, Virginia; 4Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University, Jinju, Korea; and 5Department of Surgery and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee Abstract regulatory complex, NELF-B (6). In its role as NELF-B, COBRA1 has a Cofactor of BRCA1 (COBRA1) is a newly characterized been shown to bind directly to the active estrogen receptor (ER) member of the negative elongation factor (NELF) complex. and recruit the other NELF subunits to stall RNA polymerase II at In this work, we show that COBRA1 is overexpressed in the the promoter proximal region, thereby preventing transcription majority of primary upper gastrointestinal adenocarcinomas elongation (7, 8). The polymerase stalling mechanism is used by the (UGC), and its overexpression correlates with down-regulation NELF complex to inhibit estrogen-induced transcription of TFF1 in of TFF1. We have detected overexpression of COBRA1 mRNA breast cancer (7). using quantitative real-time reverse transcription-PCR in 28 TFF1 is one of a family of highly conserved, secreted trefoil (79%) primary UGCs. Immunohistochemical analysis of UGC peptide proteins (9). TFF1 is normally expressed only in the upper gastrointestinal system, in the upper portion of the glandular pits tissue arrays that contained 70 tumor samples showed moderate-strong staining for COBRA1 in 60 (84%) tumors.
    [Show full text]
  • Overexpression of Cofactor of BRCA1 in Hepg2 Cells: a Step Towards Understanding the Role of COBRA1 in Hepatocellular Carcinoma
    American University in Cairo AUC Knowledge Fountain Theses and Dissertations 2-1-2016 Overexpression of cofactor of BRCA1 in HepG2 cells: A step towards understanding the role of COBRA1 in hepatocellular carcinoma Razan Jamil Masad Follow this and additional works at: https://fount.aucegypt.edu/etds Recommended Citation APA Citation Masad, R. (2016).Overexpression of cofactor of BRCA1 in HepG2 cells: A step towards understanding the role of COBRA1 in hepatocellular carcinoma [Master’s thesis, the American University in Cairo]. AUC Knowledge Fountain. https://fount.aucegypt.edu/etds/552 MLA Citation Masad, Razan Jamil. Overexpression of cofactor of BRCA1 in HepG2 cells: A step towards understanding the role of COBRA1 in hepatocellular carcinoma. 2016. American University in Cairo, Master's thesis. AUC Knowledge Fountain. https://fount.aucegypt.edu/etds/552 This Thesis is brought to you for free and open access by AUC Knowledge Fountain. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of AUC Knowledge Fountain. For more information, please contact [email protected]. Overexpression of Cofactor of BRCA1 in HepG2 Cells: A Step Towards Understanding the Role of COBRA1 in Hepatocellular Carcinoma A Thesis Submitted to the Biotechnology Master’s Program In partial fulfilment of the requirements for the Degree of Master of Science By: Razan Jamil Masad (Under the supervision of Dr. Asma Amleh) December/2016 The American University in Cairo School of Sciences and Engineering (SSE) Overexpression
    [Show full text]
  • Hela Cell Line, a Model to Study the Role of Cofactor of BRCA1 in Cervical Cancer
    American University in Cairo AUC Knowledge Fountain Theses and Dissertations 6-1-2018 HeLa cell line, a model to study the role of cofactor of BRCA1 in cervical cancer Noha Saad Abd Elhamied Follow this and additional works at: https://fount.aucegypt.edu/etds Recommended Citation APA Citation Abd Elhamied, N. (2018).HeLa cell line, a model to study the role of cofactor of BRCA1 in cervical cancer [Master’s thesis, the American University in Cairo]. AUC Knowledge Fountain. https://fount.aucegypt.edu/etds/1344 MLA Citation Abd Elhamied, Noha Saad. HeLa cell line, a model to study the role of cofactor of BRCA1 in cervical cancer. 2018. American University in Cairo, Master's thesis. AUC Knowledge Fountain. https://fount.aucegypt.edu/etds/1344 This Thesis is brought to you for free and open access by AUC Knowledge Fountain. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of AUC Knowledge Fountain. For more information, please contact [email protected]. School of Sciences and Engineering HELA CELL LINE, A MODEL TO STUDY THE ROLE OF COFACTOR OF BRCA1 IN CERVICAL CANCER A Thesis Submitted to the Biotechnology Master’s Program In partial fulfilment of the requirements for the Degree of Master of Science By Noha Saad Abd Elhamied Bachelor of Pharmaceutical Sciences Under the supervision of Dr. Asma Amleh Associate Professor, Department of Biology The American University in Cairo May 2018 The American University in Cairo School of Sciences and Engineering (SSE) HELA CELL LINE, A MODEL TO
    [Show full text]
  • MOL #82305 TITLE PAGE Title: Induced CYP3A4 Expression In
    Downloaded from molpharm.aspetjournals.org at ASPET Journals on September 28, 2021 1 This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted. The final version may differ from this version. This article has not been copyedited and formatted.
    [Show full text]
  • 10Th Anniversary of the Human Genome Project
    Grand Celebration: 10th Anniversary of the Human Genome Project Volume 1 Edited by John Burn, James R. Lupski, Karen E. Nelson and Pabulo H. Rampelotto Printed Edition of the Special Issue Published in Genes www.mdpi.com/journal/genes John Burn, James R. Lupski, Karen E. Nelson and Pabulo H. Rampelotto (Eds.) Grand Celebration: 10th Anniversary of the Human Genome Project Volume 1 This book is a reprint of the special issue that appeared in the online open access journal Genes (ISSN 2073-4425) in 2014 (available at: http://www.mdpi.com/journal/genes/special_issues/Human_Genome). Guest Editors John Burn University of Newcastle UK James R. Lupski Baylor College of Medicine USA Karen E. Nelson J. Craig Venter Institute (JCVI) USA Pabulo H. Rampelotto Federal University of Rio Grande do Sul Brazil Editorial Office Publisher Assistant Editor MDPI AG Shu-Kun Lin Rongrong Leng Klybeckstrasse 64 Basel, Switzerland 1. Edition 2016 MDPI • Basel • Beijing • Wuhan ISBN 978-3-03842-123-8 complete edition (Hbk) ISBN 978-3-03842-169-6 complete edition (PDF) ISBN 978-3-03842-124-5 Volume 1 (Hbk) ISBN 978-3-03842-170-2 Volume 1 (PDF) ISBN 978-3-03842-125-2 Volume 2 (Hbk) ISBN 978-3-03842-171-9 Volume 2 (PDF) ISBN 978-3-03842-126-9 Volume 3 (Hbk) ISBN 978-3-03842-172-6 Volume 3 (PDF) © 2016 by the authors; licensee MDPI, Basel, Switzerland. All articles in this volume are Open Access distributed under the Creative Commons License (CC-BY), which allows users to download, copy and build upon published articles even for commercial purposes, as long as the author and publisher are properly credited, which ensures maximum dissemination and a wider impact of our publications.
    [Show full text]
  • Wolf-Hirschhorn Syndrome-Associated Genes Are Enriched in Motile Neural Crest Cells
    bioRxiv preprint doi: https://doi.org/10.1101/471672; this version posted November 15, 2018. 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 4.0 International license. Wolf-Hirschhorn Syndrome-associated genes are enriched in motile neural crest cells and affect craniofacial development in Xenopus laevis Short Title: WHS-affected genes shape craniofacial morphology Alexandra Mills#, Elizabeth Bearce#, Rachael Cella, Seung Woo Kim, Megan Selig, Sangmook Lee, and Laura Anne Lowery* Biology Department, Boston College, Chestnut Hill, MA, USA * Corresponding author Email: [email protected] #These authors contributed equally to this work. Keywords craniofacial development | developmental disorders | 4p- | Wolf-Hirschhorn Syndrome | WHSC1 | WHSC2 | LETM1 | TACC3 | neural crest | disease model bioRxiv preprint doi: https://doi.org/10.1101/471672; this version posted November 15, 2018. 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 4.0 International license. 1 Abstract 2 Wolf-Hirschhorn Syndrome (WHS) is a human developmental disorder arising from a 3 hemizygous perturbation, typically a microdeletion, on the short arm of chromosome four. In 4 addition to pronounced intellectual disability, seizures, and delayed growth, WHS presents with 5 a characteristic facial dysmorphism and varying prevalence of microcephaly, micrognathia, 6 cartilage malformation in the ear and nose, and facial asymmetries. These affected craniofacial 7 tissues all derive from a shared embryonic precursor, the cranial neural crest, inviting the 8 hypothesis that one or more WHS-affected genes may be critical regulators of neural crest 9 development or migration.
    [Show full text]