A Neuro-Specific Hedgehog-Responsive Enhancer from Intron 1 of the Murine Laminin Alpha 1 Gene

Total Page:16

File Type:pdf, Size:1020Kb

A Neuro-Specific Hedgehog-Responsive Enhancer from Intron 1 of the Murine Laminin Alpha 1 Gene A neuro-specific hedgehog-responsive enhancer from intron 1 of the murine laminin alpha 1 gene Thesis presented for the degree of PhD by Kalin Dimitrov Narov Department of Biomedical Science University of Sheffield September 2013 i Acknowledgements I thank my supervisors Anne-Gaëlle Borycki and Philip Ingham for all expert guidance, patience, and for the opportunity to study vertebrate development in their laboratories. I also thank my advisors Pen Rashbass and Vincent Cunliffe for the helpful advices and their critical analyses on my work, and our collaborator Norris Ray Dunn for advising me on mouse transgenics and providing me with mouse embryos. Many thanks also to Shantisree Rayagiri, Joseph B. Pickering, Claire Anderson, Ashish Maurya, Weixin Niah, Harriet Jackson, Raymond Lee, Xingang Wang, Yogavali Pooblan for helping me with text formatting and embryo harvesting, and for providing me with reagents and advices. I am also grateful to the whole D-floor community, as well as to Martin Zeidler’s and Marcelo Rivolta’s labs for letting me use their equipment. Last but not least, I thank my family for the constant support and encouragement, and especially my parents for nurturing in me the love to nature and knowledge. Therefore, I dedicate this work to the memory of my father. ii Abstract Laminin alpha 1 (LAMA1) is a major component of the earliest basement membranes in the mammalian embryo. Disruption of the murine Lama1 gene result in lethal failure of germ layer differentiation and extraembryonic membrane formation at gastrulation stages, while conditional deletion of Lama1 leads to aberrant organization of retinal neurons and vasculature, and defects in cerebellar glia and granule cell precursors later in development. Similarly, inactivation of lama1 in zebrafish affects lens, retina and anterior notochord development. This diverse range of phenotypes in Lama1-deficient animals reflects the complexity of its expression pattern during embryogenesis, which is largely conserved among vertebrates. Major sites of Lama1 transcription in the mouse embryo are the neural tube, presomitic mesoderm, somites, nephrogenic mesoderm, head mesenchyme and the lens. However, little is known about the signaling mechanisms governing the spatio-temporal control of Lama1 transcription. Previous studies in our lab revealed a requirement for SHH signaling in the transcription of Lama1 in the somites and neural tube of mouse embryos. Therefore, I hypothesized that SHH might directly modulate Lama1 expression via the binding of GLI transcription factors to regulatory regions in the Lama1 locus. In this study, I identified a cis-regulatory element that may be involved in the SHH-dependent control of Lama1 expression in the murine embryo. I began my study with a phylogenetic footprinting approach that uncovered 25 conserved non-coding elements upstream of the murine Lama1 locus, some of which contained GLI binding motifs. Subsequent luciferase reporter-based analysis in cell culture with a subset of the CNEs did not provide convincing evidence for enhancer- and/or silencer- like properties of the elements, except for CNE7. The CNEs were further characterised using an in vivo transgenesis reporter screen in zebrafish, which uncovered a skeletal-muscle specific regulatory region. In parallel, a detailed survey of the existing literature revealed the presence of a non- conserved GLI-occupied region in intron 1 of the murine Lama1 gene. Subsequently, I showed that this element behaves as a tissue-specific enhancer driving reporter expression in the neural tube of mouse and zebrafish embryos. I provided evidence that active Hh signaling is required and sufficient for the activity of this enhancer. Finally, I demonstrated that the GLI binding motifs within the element are essential for its function. Altogether, these results suggest that SHH may directly control Lama1 transcription in the mouse neural tube via an intronic enhancer, and also provide further insight in the relationship between cell signaling and the regulated expression of extracellular matrix components in development and disease. iii Table of Contents Chapter 1 ................................................................................................................................................i Introduction......................................................................................................................................... 1 1. Introduction ................................................................................................................................. 2 1.1. Basement membranes and animal development................................................................................. 2 1.2. Laminin alpha 1 – gene organization, protein structure and assembly ............................................... 2 1.2.1. Laminin α1 is part of a heterotrimeric complex .......................................................................... 4 1.2.2. Interactions of Laminin-111 with other ECM components and cellular receptors ..................... 6 1.2.3. Laminin-111 and basement membrane assembly ....................................................................... 7 1.3. Expression of laminin α 1 .................................................................................................................. 7 1.3.1. Lama1 expression in the mouse and human ............................................................................... 8 1.3.2. Lama1 expression in zebrafish.................................................................................................... 8 1.4. Regulation of Lama1 expression ........................................................................................................ 9 1.4.1. Signalling pathways and control of Lama1 expression in vitro .................................................. 9 1.4.2. cis-regulatory elements in Lama1 transcription ........................................................................ 11 1.4.3. Regulation of Lama1 transcription in embryonic development ................................................ 14 1.5. Laminin expression in skeletal muscle development ....................................................................... 14 1.6. SHH is required for Lama1 expression and myotomal basement membrane assembly ................... 16 1.6.1. Shh-/- mutant mouse embryos fail to express Lama1 in the somites and neural tube ................ 16 1.6.2. Shh is required for lama1 expression in zebrafish .................................................................... 17 1.7. Functions of laminin α1 in development .......................................................................................... 17 1.7.1. Laminins and neural development ............................................................................................ 17 1.7.1.1. Laminins and neural stem cells ......................................................................................... 17 1.7.1.2. Laminins and cortical morphogenesis ............................................................................... 18 1.7.1.3. Laminin α1 in axonal growth and migration ..................................................................... 19 1.7.2. Laminins and neural crest cells ................................................................................................. 20 1.7.3. Laminin α1 in ocular development ........................................................................................... 21 1.7.4. Laminin α1 and kidney development ........................................................................................ 22 1.7.5. Laminin α1 in skeletal muscle development ............................................................................. 22 1.8. The SHH signaling pathway ............................................................................................................ 23 1.8.1.Expression, secretion and diffusion of Shh ................................................................................ 24 1.8.2. Transduction of the HH signal .................................................................................................. 25 1.8.3. The GLI transcription factors are mediators of HH signaling in vertebrates ............................ 26 1.8.4. SHH and dorso-ventral patterning of the neural tube ............................................................... 28 1.9. Transcription-regulatory elements…………………………………………………………………30 1.9.1. The core promoter……………………………………………………………………………..30 1.9.2. Transcriptional enhancers……………………………………………………………………..31 1.9.3. Transcriptional silencers………………………………………………………………………32 1.9.4. Insulators………………………………………………………………………………………32 1.10. Methods for the prediction of transcription-regulatory elements………………………………...33 1.10.1. Direct (biochemical) methods………………………………………………………………..33 1.10.1.1. Prediction of TREs based on transcription factor occupancy and/or histone modifications as revealed by ChIP-seq………………………………………………………………………………………….33 1.10.1.2. Prediction of TREs in DNaseI hypersensitive domains…………………………………35 1.10.2. Indirect methods……………………………………………………………………………...36 1.10.2.1. Computational prediction of TREs based on sequence organisation/pattern, irrespective of overall conservation…………………………………………………………………………………………...36 iv 1.10.2.2. Comparative genomics and TRE prediction…………………………………………….38 1.11. Functional validation of candidate transcription-regulatory elements……………………………39 1.11.1. Zebrafish as a tool for the validation
Recommended publications
  • Impact of ZBTB7A Hypomethylation and Expression Patterns on Treatment Response to Hydroxyurea Vasiliki Chondrou1†, Eleana F
    Chondrou et al. Human Genomics (2018) 12:45 https://doi.org/10.1186/s40246-018-0177-z PRIMARY RESEARCH Open Access Impact of ZBTB7A hypomethylation and expression patterns on treatment response to hydroxyurea Vasiliki Chondrou1†, Eleana F. Stavrou1†, Georgios Markopoulos2, Alexandra Kouraklis-Symeonidis3, Vasilios Fotopoulos4, Argiris Symeonidis5, Efthymia Vlachaki6, Panagiota Chalkia7, George P. Patrinos8, Adamantia Papachatzopoulou9 and Argyro Sgourou1* Abstract Background: We aimed to clarify the emerging epigenetic landscape in a group of genes classified as “modifier genes” of the β-type globin genes (HBB cluster), known to operate in trans to accomplish the two natural developmental switches in globin expression, from embryonic to fetal during the first trimester of conception and from fetal to adult around the time of birth. The epigenetic alterations were determined in adult sickle cell anemia (SCA) homozygotes and SCA/β-thalassemia compound heterozygotes of Greek origin, who are under hydroxyurea (HU) treatment. Patients were distinguished in HU responders and HU non-responders (those not benefited from the HU) and both, and in vivo and in vitro approaches were implemented. Results: We examined the CpG islands’ DNA methylation profile of BCL11A, KLF1, MYB, MAP3K5, SIN3A, ZBTB7A,andGATA2, along with γ-globin and LRF/ZBTB7A expression levels. In vitro treatment of hematopoietic stem cells (HSCs) with HU induced a significant DNA hypomethylation pattern in ZBTB7A (p*, 0.04) and GATA2 (p*, 0.03) CpGs exclusively in the HU non-responders. Also, this group of patients exhibited significantly elevated baseline methylation patterns in ZBTB7A, before the HU treatment, compared to HU responders (p*, 0.019) and to control group of healthy individuals (p*, 0.021) , which resembles a potential epigenetic barrier for the γ-globin expression.
    [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]
  • Prox1regulates the Subtype-Specific Development of Caudal Ganglionic
    The Journal of Neuroscience, September 16, 2015 • 35(37):12869–12889 • 12869 Development/Plasticity/Repair Prox1 Regulates the Subtype-Specific Development of Caudal Ganglionic Eminence-Derived GABAergic Cortical Interneurons X Goichi Miyoshi,1 Allison Young,1 Timothy Petros,1 Theofanis Karayannis,1 Melissa McKenzie Chang,1 Alfonso Lavado,2 Tomohiko Iwano,3 Miho Nakajima,4 Hiroki Taniguchi,5 Z. Josh Huang,5 XNathaniel Heintz,4 Guillermo Oliver,2 Fumio Matsuzaki,3 Robert P. Machold,1 and Gord Fishell1 1Department of Neuroscience and Physiology, NYU Neuroscience Institute, Smilow Research Center, New York University School of Medicine, New York, New York 10016, 2Department of Genetics & Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, 3Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan, 4Laboratory of Molecular Biology, Howard Hughes Medical Institute, GENSAT Project, The Rockefeller University, New York, New York 10065, and 5Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724 Neurogliaform (RELNϩ) and bipolar (VIPϩ) GABAergic interneurons of the mammalian cerebral cortex provide critical inhibition locally within the superficial layers. While these subtypes are known to originate from the embryonic caudal ganglionic eminence (CGE), the specific genetic programs that direct their positioning, maturation, and integration into the cortical network have not been eluci- dated. Here, we report that in mice expression of the transcription factor Prox1 is selectively maintained in postmitotic CGE-derived cortical interneuron precursors and that loss of Prox1 impairs the integration of these cells into superficial layers. Moreover, Prox1 differentially regulates the postnatal maturation of each specific subtype originating from the CGE (RELN, Calb2/VIP, and VIP).
    [Show full text]
  • Cell Reprogramming Technologies for Treatment And
    CELL REPROGRAMMING TECHNOLOGIES FOR TREATMENT AND UNDERSTANDING OF GENETIC DISORDERS OF MYELIN by ANGELA MARIE LAGER Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy Thesis advisor: Paul J Tesar, PhD Department of Genetics and Genome Sciences CASE WESTERN RESERVE UNIVERSITY May 2015 CASE WESTERN RESERVE UNIVERSITY SCHOOL OF GRADUATE STUDIES We hereby approve the thesis/dissertation of Angela Marie Lager Candidate for the Doctor of Philosophy degree*. (signed) Ronald A Conlon, PhD (Committee Chair) Paul J Tesar, PhD (Advisor) Craig A Hodges, PhD Warren J Alilain, PhD (date) 31 March 2015 *We also certify that written approval has been obtained from any proprietary material contained therein. TABLE OF CONTENTS Table of Contents……………………………………………………………………….1 List of Figures……………………………………………………………………………4 Acknowledgements……………………………………………………………………..7 Abstract…………………………………………………………………………………..8 Chapter 1: Introduction and Background………………………………………..11 1.1 Overview of mammalian oligodendrocyte development in the spinal cord and myelination of the central nervous system…………………..11 1.1.1 Introduction……………………………………………………..11 1.1.2 The establishment of the neuroectoderm and ventral formation of the neural tube…………………………………..12 1.1.3 Ventral patterning of the neural tube and specification of the pMN domain in the spinal cord……………………………….15 1.1.4 Oligodendrocyte progenitor cell production through the process of gliogenesis ………………………………………..16 1.1.5 Oligodendrocyte progenitor cell to oligodendrocyte differentiation…………………………………………………...22
    [Show full text]
  • Viewed in [2, 3])
    Yildiz et al. Neural Development (2019) 14:5 https://doi.org/10.1186/s13064-019-0129-x RESEARCH ARTICLE Open Access Zebrafish prdm12b acts independently of nkx6.1 repression to promote eng1b expression in the neural tube p1 domain Ozge Yildiz1, Gerald B. Downes2 and Charles G. Sagerström1* Abstract Background: Functioning of the adult nervous system depends on the establishment of neural circuits during embryogenesis. In vertebrates, neurons that make up motor circuits form in distinct domains along the dorsoventral axis of the neural tube. Each domain is characterized by a unique combination of transcription factors (TFs) that promote a specific fate, while repressing fates of adjacent domains. The prdm12 TF is required for the expression of eng1b and the generation of V1 interneurons in the p1 domain, but the details of its function remain unclear. Methods: We used CRISPR/Cas9 to generate the first germline mutants for prdm12 and employed this resource, together with classical luciferase reporter assays and co-immunoprecipitation experiments, to study prdm12b function in zebrafish. We also generated germline mutants for bhlhe22 and nkx6.1 to examine how these TFs act with prdm12b to control p1 formation. Results: We find that prdm12b mutants lack eng1b expression in the p1 domain and also possess an abnormal touch-evoked escape response. Using luciferase reporter assays, we demonstrate that Prdm12b acts as a transcriptional repressor. We also show that the Bhlhe22 TF binds via the Prdm12b zinc finger domain to form a complex. However, bhlhe22 mutants display normal eng1b expression in the p1 domain. While prdm12 has been proposed to promote p1 fates by repressing expression of the nkx6.1 TF, we do not observe an expansion of the nkx6.1 domain upon loss of prdm12b function, nor is eng1b expression restored upon simultaneous loss of prdm12b and nkx6.1.
    [Show full text]
  • CNS Myelin Paranodes Require Nkx6-2 Homeoprotein Transcriptional Activity for Normal Structure
    The Journal of Neuroscience, December 15, 2004 • 24(50):11215–11225 • 11215 Development/Plasticity/Repair CNS Myelin Paranodes Require Nkx6-2 Homeoprotein Transcriptional Activity for Normal Structure Cherie Southwood,1,2 Chris He,1 James Garbern,2,4 John Kamholz,2,4 Edgardo Arroyo,5 and Alexander Gow1,2,3,4 1Brookdale Center for Molecular Biology, Mount Sinai School of Medicine, New York, New York, 10029, 2Center for Molecular Medicine and Genetics, 3Carman and Ann Adams Department of Pediatrics, 4Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, 48201, and 5Department of Neurology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania, 19104 Homeodomain proteins play critical roles during development in cell fate determination and proliferation, but few studies have defined gene regulatory networks for this class of transcription factors in differentiated cells. Using a lacZ-knock-in strategy to ablate Nkx6-2, we find that the Nkx6-2 promoter is active embryonically in neuroblasts and postnatally in oligodendrocytes. In addition to neurological deficits, we find widespread ultrastructural abnormalities in CNS white matter and aberrant expression of three genes encoding a paranodal microtubule destabilizing protein, stathmin 1, and the paranodal cell adhesion molecules neurofascin and contactin. The involvement of these downstream proteins in cytoskeletal function and cell adhesion reveals mechanisms whereby Nkx6-2 directly or indirectly regulates axon–glial interactions at myelin paranodes. Nkx6-2 does not appear to be the central regulator of axoglial junction assembly; nonetheless, our data constitute the first evidence of such a regulatory network and provide novel insights into the mechanism and effector molecules that are involved.
    [Show full text]
  • The BTB-ZF Family of Transcription Factors: Key Regulators of Lineage
    The BTB-ZF Family of Transcription Factors: Key Regulators of Lineage Commitment and Effector Function Development in the Immune System This information is current as of September 27, 2021. Aimee M. Beaulieu and Derek B. Sant'Angelo J Immunol 2011; 187:2841-2847; ; doi: 10.4049/jimmunol.1004006 http://www.jimmunol.org/content/187/6/2841 Downloaded from References This article cites 103 articles, 44 of which you can access for free at: http://www.jimmunol.org/content/187/6/2841.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on September 27, 2021 *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 © 2011 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The BTB-ZF Family of Transcription Factors: Key Regulators of Lineage Commitment and Effector Function Development in the Immune System Aimee M. Beaulieu* and Derek B. Sant’Angelo*,†,‡ Successful immunity depends upon the activity of mul- 1, -2, -4, -5, and -7 (1–11).
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • A Population-Specific Major Allele Reference Genome from the United
    Edith Cowan University Research Online ECU Publications Post 2013 2021 A population-specific major allele efr erence genome from the United Arab Emirates population Gihan Daw Elbait Andreas Henschel Guan K. Tay Edith Cowan University Habiba S. Al Safar Follow this and additional works at: https://ro.ecu.edu.au/ecuworkspost2013 Part of the Life Sciences Commons, and the Medicine and Health Sciences Commons 10.3389/fgene.2021.660428 Elbait, G. D., Henschel, A., Tay, G. K., & Al Safar, H. S. (2021). A population-specific major allele reference genome from the United Arab Emirates population. Frontiers in Genetics, 12, article 660428. https://doi.org/10.3389/ fgene.2021.660428 This Journal Article is posted at Research Online. https://ro.ecu.edu.au/ecuworkspost2013/10373 fgene-12-660428 April 19, 2021 Time: 16:18 # 1 ORIGINAL RESEARCH published: 23 April 2021 doi: 10.3389/fgene.2021.660428 A Population-Specific Major Allele Reference Genome From The United Arab Emirates Population Gihan Daw Elbait1†, Andreas Henschel1,2†, Guan K. Tay1,3,4,5 and Habiba S. Al Safar1,3,6* 1 Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates, 2 Department of Electrical Engineering and Computer Science, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates, 3 Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates, 4 Division of Psychiatry, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, Australia, 5 School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia, 6 Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates The ethnic composition of the population of a country contributes to the uniqueness of each national DNA sequencing project and, ideally, individual reference genomes are required to reduce the confounding nature of ethnic bias.
    [Show full text]
  • SUPPLEMENTARY NOTE Co-Activation of GR and NFKB
    SUPPLEMENTARY NOTE Co-activation of GR and NFKB alters the repertoire of their binding sites and target genes. Nagesha A.S. Rao1*, Melysia T. McCalman1,*, Panagiotis Moulos2,4, Kees-Jan Francoijs1, 2 2 3 3,5 Aristotelis Chatziioannou , Fragiskos N. Kolisis , Michael N. Alexis , Dimitra J. Mitsiou and 1,5 Hendrik G. Stunnenberg 1Department of Molecular Biology, Radboud University Nijmegen, the Netherlands 2Metabolic Engineering and Bioinformatics Group, Institute of Biological Research and Biotechnology, National Hellenic Research Foundation, Athens, Greece 3Molecular Endocrinology Programme, Institute of Biological Research and Biotechnology, National Hellenic Research Foundation, Greece 4These authors contributed equally to this work 5 Corresponding authors E-MAIL: [email protected] ; TEL: +31-24-3610524; FAX: +31-24-3610520 E-MAIL: [email protected] ; TEL: +30-210-7273741; FAX: +30-210-7273677 Running title: Global GR and NFKB crosstalk Keywords: GR, p65, genome-wide, binding sites, crosstalk SUPPLEMENTARY FIGURES/FIGURE LEGENDS AND SUPPLEMENTARY TABLES 1 Rao118042_Supplementary Fig. 1 A Primary transcript Mature mRNA TNF/DMSO TNF/DMSO 8 12 r=0.74, p< 0.001 r=0.61, p< 0.001 ) 2 ) 10 2 6 8 4 6 4 2 2 0 Fold change (mRNA) (log Fold change (primRNA) (log 0 −2 −2 −2 0 2 4 −2 0 2 4 Fold change (RNAPII) (log2) Fold change (RNAPII) (log2) B chr5: chrX: 56 _ 104 _ DMSO DMSO 1 _ 1 _ 56 _ 104 _ TA TA 1 _ 1 _ 56 _ 104 _ TNF TNF Cluster 1 1 _ Cluster 2 1 _ 56 _ 104 _ TA+TNF TA+TNF 1 _ 1 _ CCNB1 TSC22D3 chr20: chr17: 25 _ 33 _ DMSO DMSO 1 _ 1 _ 25 _ 33 _ TA TA 1 _ 1 _ 25 _ 33 _ TNF TNF Cluster 3 1 _ Cluster 4 1 _ 25 _ 33 _ TA+TNF TA+TNF 1 _ 1 _ GPCPD1 CCL2 chr6: chr22: 77 _ 35 _ DMSO DMSO 1 _ 77 _ 1 _ 35 _ TA TA 1 _ 1 _ 77 _ 35 _ TNF Cluster 5 Cluster 6 TNF 1 _ 1 _ 77 _ 35 _ TA+TNF TA+TNF 1 _ 1 _ TNFAIP3 DGCR6 2 Supplementary Figure 1.
    [Show full text]
  • An All-To-All Approach to the Identification of Sequence-Specific Readers for Epigenetic DNA Modifications on Cytosine
    bioRxiv preprint doi: https://doi.org/10.1101/638700; this version posted May 16, 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. An All-to-All Approach to the Identification of Sequence-Specific Readers for Epigenetic DNA Modifications on Cytosine Guang Song1,6, Guohua Wang2,6, Ximei Luo2,3,6, Ying Cheng4, Qifeng Song1, Jun Wan3, Cedric Moore1, Hongjun Song5, Peng Jin4, Jiang Qian3,7,*, Heng Zhu1,7,8,* 1Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 2School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China 3Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 4Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA 5Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA 19104, USA 6These authors contributed equally 7Senior author 8Lead Contact *Correspondence: [email protected] (H.Z.), [email protected] (J.Q.). 1 bioRxiv preprint doi: https://doi.org/10.1101/638700; this version posted May 16, 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. SUMMARY Epigenetic modifications of DNA in mammals play important roles in many biological processes. Identification of readers of these epigenetic marks is a critical step towards understanding the underlying molecular mechanisms. Here, we report the invention and application of an all-to-all approach, dubbed Digital Affinity Profiling via Proximity Ligation (DAPPL), to simultaneously profile human TF-DNA interactions using mixtures of random DNA libraries carrying four different epigenetic modifications (i.e., 5-methylcytosine, 5- hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine).
    [Show full text]
  • Genetic Determinants of Inter-Individual Variations in DNA Methylation
    Genetic determinants of inter-individual variations in DNA methylation Dissertation zur Erlangung des Doktorgrades der Naturwissenschaften (Dr. rer. nat.) der Fakultät für Biologie und Vorklinische Medizin der Universität Regensburg vorgelegt von Julia Wimmer (geb. Wegner) aus Neubrandenburg im Jahr 2014 The present work was carried out at the Clinic and Polyclininc of Internal Medicine III at the University Hospital Regensburg from March 2010 to August 2014. Die vorliegende Arbeit entstand im Zeitraum von März 2010 bis August 2014 an der Klinik und Poliklinik für Innere Medizin III des Universitätsklinikums Regensburg. Das Promotionsgesuch wurde eingereicht am: 01. September 2014 Die Arbeit wurde angeleitet von: Prof. Dr. Michael Rehli Prüfungsausschuss: Vorsitzender: Prof. Dr. Herbert Tschochner Erstgutachter: Prof. Dr. Michael Rehli Zweitgutachter: Prof. Dr. Axel Imhof Drittprüfer: Prof. Dr. Gernot Längst Ersatzprüfer: PD Dr. Joachim Griesenbeck Unterschrift: ____________________________ Who seeks shall find. (Sophocles) Table of Contents TABLE OF CONTENTS .......................................................................................................................... I LIST OF FIGURES ................................................................................................................................ IV LIST OF TABLES ................................................................................................................................... V 1 INTRODUCTION ..........................................................................................................................
    [Show full text]