Characterizing the Effect of Myh7b ASO Treatment in Cardiomyocytes Maturation in Cella

Total Page:16

File Type:pdf, Size:1020Kb

Characterizing the Effect of Myh7b ASO Treatment in Cardiomyocytes Maturation in Cella Characterizing the Effect of Myh7b ASO Treatment in Cardiomyocytes Maturation In Cella Jose Santos Navarro Arriola Molecular, Cellular, and Developmental Biology Departmental Honors Thesis University of Colorado Boulder Defense Date: April 7th, 2021 Thesis Advisor: Dr. Leslie Leinwand (MCDB & BioFrontiers Institute) Honors Council Representative: Dr. Joel KralJ (MCDB) Outside Department Advisor: Member: Dr. Katharine Semsar (Arts & Sciences) Abstract Myh7b is an ancient gene encoding a Myosin Heavy Chain (MyHC) protein that has been repurposed for alternative regulatory roles in cardiomyocytes due to an alternative spliced exon skipping mechanism. To better understand the role of Myh7b gene in cardiomyocytes, this project investigated cardiomyocytes differentiation and maturation. The goal was to expand our knowledge of Myh7b’s roles in cardiomyocytes. A study prior to this project showed that the prominent gene product candidate of the Myh7b gene was the long non-coding RNA (lncMyh7b) produced by the exon-skipping mechanism. This was achieved by eliminating miR-499 and Myh7b short peptide (Myh7b_sp) as candidates. A portion of this project focused on the western blotting for Myh7b_sp, which was found to be undetectable in the human heart. A CRISPR knockout system was developed, but an ASO knockdown system was pursued instead for reducing lncMyh7b as it proved more versatile for experimental design. The lncMyh7b ASO treated hiPSCs showed a heterogenous phenotype, in contrast to the control ASO treated cells which successfully differentiated into cardiomyocytes. Only one well sample of lncMyh7b ASO showed signs of delayed differentiation, whereas all of the control ASO cells followed the established cardiomyocyte differentiation timeline. Along with previous studies. the data of this project points to lncMyh7b ASO treatment disrupting normal cardiomyocyte differentiation and maturation. Specifically, we hypothesiZed that lncMyh7b may affect the Wnt pathway, as temporal control of Wnt is essential for successful differentiation. Keywords: Myh7b, lncMyh7b, Myh7b_sp, Anti Sense Oligos, hiPSC, Wnt Introduction Proper heart development and maturation is dictated by finely-tuned regulations in sarcomeric proteins like those from the myosin family of motor proteins involved in heart contraction. Furthermore, one of the hallmarks of human heart disease are changes to the myosin ratio between the two most common myosin heavy chain (MyHC) proteins: β-MyHC and α- MyHC. In the human heart, β-MyHC comprises 90% of MyHC proteins and α-MyHC makes up the remaining 10% (Nakao et al. 1997). These two MyHC proteins’ differences in speed and energy efficiency contribute to different properties of the sarcomeric unit’s contractility. Therefore, changes to the β-MyHC/α-MyHC ratio has drastic consequences on the heart overall. The third MyHC gene expressed in the heart is called Myh7b, which is predicted to have a slow- twitch property like β-MyHC. However, Myh7b protein is not produced in the heart, due to a post-transcriptional exon-skipping mechanism that prevents translation of the RNA. Despite this, knockdown of Myh7b has been shown to reduce β-MyHC protein expression (Broadwell et al. 2020 Sep 6). Myh7b has a gene product with some regulatory role with the β-MyHC/α-MyHC ratio While previous research with Myh7b has used in vivo mice models, mice have the opposite ratio of β-MyHC/α-MyHC proteins (Krenz and Robbins 2004), and therefore we decided to use human induced pluripotent stem cells (hiPSCs) to examine this regulation. hiPSCs better pertain to human specific regulatory networks. The focus of previous hiPSCs projects looked into the role of acute changes of Myh7b expression in mature cardiomyocytes. No research has explored in the role of Myh7b during cardiomyocyte differentiation and maturation process. This is a perfect opportunity to further shed light on the role of Myh7b in cardiomyocytes. Myh7b is an ancient myosin gene that has been repurposed for novel regulatory roles outside of the typical protein interactions, and currently the gene encodes an RNA mediated regulation in cardiomyocyte. We hypothesiZe that this regulation may play some role in cardiomyocyte differentiation and maturation. My project hopes to explore this possibility. Background Cardiomyocytes Cardiomyocytes (CMs) are the terminally differentiated cells that comprise the part of the heart tissue that provide contractile force. This is because of sarcomeric units pulling in on each other through electrophysiological work by Ca2+ exchange. When millions of sarcomeric units work in unison, the tissue level contractions provide enough force generation to keep the heart beating and blood pumped throughout the organism. The sarcomere is comprised of repeating units of different structural and enzymatic proteins including myosin proteins. Myosin heavy chain (MyHC) proteins are motor proteins that convert chemical energy into kinetic energy. The sarcomeric MyHC proteins involved in contraction are a part of a hexameric complex. Two heavy chains will dimeriZe proteins via their rod domains, which will further self-assemble into the thick filament, leaving the ATPase head domains free to pull on the actin thin filament. Each heavy chain is further decorated by two regulatory myosin light chains. Human cardiomyocytes express two isoforms of MyHC protein, β-MyHC (Myh7 gene) and α-MyHC (Myh6 gene). In humans, 90% of the heart’s myosin protein are β-MyHC and the remaining 10% are α-MyHC (Nakao et al. 1997). This β-MyHC/α-MyHC ratio is maintained in healthy hearts but in human heart failure there is a total shift to 100% β-MyHC. This dysregulation in the ratio exacerbates the toll on the heart (Nakao et al. 1997). Myosin Heavy Chain 7b Myosin Heavy Chain 7b (Myh7b) is another slow twitch and energy efficient MyHC isoform similar to β-MyHC. Full length Myh7b is expressed in human brain, inner ear tissues, and specialiZed muscles but is the primary cardiac myosin in chickens and reptiles (Bell et al. 2010). In human hearts, no full length Myh7b protein can be detected because of an exon-skipping mechanism relegating the transcript to become non-coding for full length protein translation, but Myh7b RNA expression was present and correlated with β-MyHC protein expression (Broadwell et al. 2020 Sep 6). The Myh7b locus has the potential to produce multiple products of interest due to an exon-skipping mechanism. We will focus on the three that can be produced by the exon-skipped transcript, as there is no evidence of full-length transcript in the heart. One of the products is an intronic miRNA called miR-499 cut from Myh7b pre-mRNA transcript intron 19. miRNAs are class of small non-coding RNAs that can have cis and trans regulation of genes. Unlike the corresponding miRNAs from β-MyHC and α-MyHC genes that did have regulatory effects on the β-MyHC/α-MyHC ratio in a mouse model (van RooiJ et al. 2009), miR- 499 does not regulate β-MyHC expression in human cardiomyocytes (Broadwell et al. 2020 Sep 6). The second product is a 27 kDa theoretical alternatively spliced Myh7b short peptide (Myh7b_sp) that can be produced by the shifted open reading frame (ORF) created by the exon skipping mechanism. In contrast to protein prediction, this project showed there were no detectable levels of Myh7b_sp in human hearts and the corresponding RNA transcript is non- coding at least in the human heart. The final product is the long non-coding Myh7b RNA (lncMyh7b) transcript with exon 8 skipped (lncMyh7b) based on current annotation. Previous research has referred the skipping mechanism to exon 7, but the rest of the data remains valid (Bell et al. 2010). lncRNAs can have cis regulatory roles in neighboring coding genes (Batista and Chang 2013; EngreitZ et al. 2016) and trans regulation on more distant genes (Nagano et al. 2008; Batista and Chang 2013). Anti-Sense Oligonucleotides Anti-Sense Oligonucleotides (ASO) are short single stranded RNA that attaches to target mRNAs through Watson-Crick pairing. After attachment, the RNA duplex is marked for degradation by RNase H activity, stall further transcription, or stop translation through steric hindrance to the ribosome (Cavagnari 2011; Kole et al. 2012). ASO treatment is a transient, selective mRNA knockdown that modulates the target gene’s mRNA concentration and, moreover, affects protein expression. The benefits of using ASO is that it is cheap, vector-less, high modularity, and appropriate for studying essential proteins due to transient inhibition. CRISPR KnocKout Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) system is a form of targeted genome editing that uses a Cas9 endonuclease with a single guided RNA (sgRNA). The sgRNA is composed of a scaffold structure and the 20-mer spacer sequence that will bind to the target sequence adjacent to a PAM sequence (ngg). After finding the target sequence, the CRISPR system makes a double stranded break (DSB). In response to the break, the cell will attempt to repair through non-homologous end joining (NHEJ) or use a template (sister chromosome or template plasmid) for homology directed repair (HDR). Knockouts occurs through frame shifts in the open reading frame caused by indels. Two near breaks in the DNA will “cutout” the region and could completely disrupt the targeted gene. Specific Aims Question: How is Myh7b expression involved in cardiomyocyte maturation? The aim of this project is to further understand the contemporary role of the ancient Myh7b gene in cardiomyocyte differentiation and maturation. The collected data will help to answer the three aims set by the project and provide more knowledge on Myh7b and potential therapeutics to cardiac afflictions caused by dysregulated β-MyHC/α-MyHC ratio. Aim 1: What gene products from Myh7b are involved in mature cardiomyocytes? Aim 2: Is Myh7b expression essential to normal cardiomyocyte maturation? Aim 3: Is Myh7b expression involved in the Wnt pathway crucial for cardiomyocyte maturation? Materials and Methods Whole Cell Lysates & Western Blotting Western blotting was used to investigate the presence of the Myh7b short peptide, one of the three gene products of Myh7b, from human tissue samples.
Recommended publications
  • Defining Functional Interactions During Biogenesis of Epithelial Junctions
    ARTICLE Received 11 Dec 2015 | Accepted 13 Oct 2016 | Published 6 Dec 2016 | Updated 5 Jan 2017 DOI: 10.1038/ncomms13542 OPEN Defining functional interactions during biogenesis of epithelial junctions J.C. Erasmus1,*, S. Bruche1,*,w, L. Pizarro1,2,*, N. Maimari1,3,*, T. Poggioli1,w, C. Tomlinson4,J.Lees5, I. Zalivina1,w, A. Wheeler1,w, A. Alberts6, A. Russo2 & V.M.M. Braga1 In spite of extensive recent progress, a comprehensive understanding of how actin cytoskeleton remodelling supports stable junctions remains to be established. Here we design a platform that integrates actin functions with optimized phenotypic clustering and identify new cytoskeletal proteins, their functional hierarchy and pathways that modulate E-cadherin adhesion. Depletion of EEF1A, an actin bundling protein, increases E-cadherin levels at junctions without a corresponding reinforcement of cell–cell contacts. This unexpected result reflects a more dynamic and mobile junctional actin in EEF1A-depleted cells. A partner for EEF1A in cadherin contact maintenance is the formin DIAPH2, which interacts with EEF1A. In contrast, depletion of either the endocytic regulator TRIP10 or the Rho GTPase activator VAV2 reduces E-cadherin levels at junctions. TRIP10 binds to and requires VAV2 function for its junctional localization. Overall, we present new conceptual insights on junction stabilization, which integrate known and novel pathways with impact for epithelial morphogenesis, homeostasis and diseases. 1 National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK. 2 Computing Department, Imperial College London, London SW7 2AZ, UK. 3 Bioengineering Department, Faculty of Engineering, Imperial College London, London SW7 2AZ, UK. 4 Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK.
    [Show full text]
  • A Missense Mutation in the RSRSP Stretch of Rbm20 Causes Dilated
    www.nature.com/scientificreports OPEN A missense mutation in the RSRSP stretch of Rbm20 causes dilated cardiomyopathy and atrial fbrillation in mice Kensuke Ihara1,2*, Tetsuo Sasano2, Yuichi Hiraoka3, Marina Togo‑Ohno4, Yurie Soejima5, Motoji Sawabe5, Megumi Tsuchiya6, Hidesato Ogawa6, Tetsushi Furukawa1 & Hidehito Kuroyanagi4* Dilated cardiomyopathy (DCM) is a fatal heart disease characterized by left ventricular dilatation and cardiac dysfunction. Recent genetic studies on DCM have identifed causative mutations in over 60 genes, including RBM20, which encodes a regulator of heart‑specifc splicing. DCM patients with RBM20 mutations have been reported to present with more severe cardiac phenotypes, including impaired cardiac function, atrial fbrillation (AF), and ventricular arrhythmias leading to sudden cardiac death, compared to those with mutations in the other genes. An RSRSP stretch of RBM20, a hotspot of missense mutations found in patients with idiopathic DCM, functions as a crucial part of its nuclear localization signals. However, the relationship between mutations in the RSRSP stretch and cardiac phenotypes has never been assessed in an animal model. Here, we show that Rbm20 mutant mice harboring a missense mutation S637A in the RSRSP stretch, mimicking that in a DCM patient, demonstrated severe cardiac dysfunction and spontaneous AF and ventricular arrhythmias mimicking the clinical state in patients. In contrast, Rbm20 mutant mice with frame‑shifting deletion demonstrated less severe phenotypes, although loss of RBM20‑dependent alternative splicing was indistinguishable. RBM20S637A protein cannot be localized to the nuclear speckles, but accumulated in cytoplasmic, perinuclear granule‑like structures in cardiomyocytes, which might contribute to the more severe cardiac phenotypes. Dilated cardiomyopathy (DCM) is a fatal cardiac disease characterized by enlargement of the cardiac chambers and impaired systolic function1.
    [Show full text]
  • Insights from Conventional and Unconventional Myosins A
    Structure-Function Analysis of Motor Proteins: Insights from Conventional and Unconventional Myosins A Thesis SUBMITTED TO THE FACULTY OF UNIVERSITY OF MINNESOTA BY Karl J. Petersen IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY Margaret A. Titus, Advisor David D. Thomas, Co-Advisor December 2016 © Karl J. Petersen 2016 Acknowledgements This thesis would not exist without the patient support of my advisors, Meg Titus and Dave Thomas. Any shortcomings are my own. Collaborators Holly Goodson, Anne Houdusse, and Gant Luxton also provided invaluable training and support. I am also grateful for essential services provided by departmental staff: Sarah Blakely Anderson, Octavian Cornea, Sarah Dittrich, Karen Hawkinson, Michelle Lewis, Mary Muwahid, Laurie O’Neill, Darlene Toedter, with apologies to others not listed. Thanks to friends and colleagues at the University of Minnesota: Ashley Arthur, Kelly Bower, Brett Colson, Sinziana Cornea, Chi Meng Fong, Greg Gillispie, Piyali Guhathakurta, Tejas Gupte, Tom Hays, Norma Jiménez Ramírez, Dawn Lowe, Allison MacLean, Santiago Martínez Cifuentes, Jared Matzke, Megan McCarthy, Joachim Mueller, Joe Muretta, Kurt Peterson, Mary Porter, Ewa Prochniewicz, Mike Ritt, Cosmo Saunders, Shiv Sivaramakrishnan, Ruth Sommese, Doug Tritschler, Brian Woolums. i Abstract Myosin motor proteins play fundamental roles in a multitude of cellular processes. Myosin generates force on cytoskeletal actin filaments to control cell shape, most dramatically during cytokinesis, and has a conserved role in defining cell polarity. Myosin contracts the actin cytoskeleton, ensuring prompt turnover of cellular adhesion sites, retracting the cell body during migration and development, and contracting muscle among diverse other functions. How myosins work, and why force generation is essential for their function, is in many cases an open question.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Expression of Normally Repressed Myosin Heavy Chain 7B in the Mammalian Heart Induces Dilated Cardiomyopathy Angela K
    ORIGINAL RESEARCH Expression of Normally Repressed Myosin Heavy Chain 7b in the Mammalian Heart Induces Dilated Cardiomyopathy Angela K. Peter, PhD;* Alberto C. Rossi, PhD;* Massimo Buvoli, MD, PhD; Christopher D. Ozeroff, BA; Claudia Crocini, PhD; Amy R. Perry, BS; Ada E. Buvoli, BS; Lindsey A. Lee, BS; Leslie A. Leinwand, PhD Background-—In mammals, muscle contraction is controlled by a family of 10 sarcomeric myosin motors. The expression of one of its members, MYH7b, is regulated by alternative splicing, and while the protein is restricted to specialized muscles such as extraocular muscles or muscle spindles, RNA that cannot encode protein is expressed in most skeletal muscles and in the heart. Remarkably, birds and snakes express MYH7b protein in both heart and skeletal muscles. This observation suggests that in the mammalian heart, the motor activity of MYH7b may only be needed during development since its expression is prevented in adult tissue, possibly because it could promote disease by unbalancing myocardial contractility. Methods and Results-—We have analyzed MYH7b null mice to determine the potential role of MYH7b during cardiac development and also generated transgenic mice with cardiac myocyte expression of MYH7b protein to measure its impact on cardiomyocyte function and contractility. We found that MYH7b null mice are born at expected Mendelian ratios and do not have a baseline cardiac phenotype as adults. In contrast, transgenic cardiac MYH7b protein expression induced early cardiac dilation in males with significantly increased left ventricular mass in both sexes. Cardiac dilation is progressive, leading to early cardiac dysfunction in males, but later dysfunction in females.
    [Show full text]
  • Effector Gene Expression Potential to Th17 Cells by Promoting Microrna
    Downloaded from http://www.jimmunol.org/ by guest on September 26, 2021 is online at: average * The Journal of Immunology published online 17 May 2013 from submission to initial decision 4 weeks from acceptance to publication http://www.jimmunol.org/content/early/2013/05/17/jimmun ol.1300351 MicroRNA-155 Confers Encephalogenic Potential to Th17 Cells by Promoting Effector Gene Expression Ruozhen Hu, Thomas B. Huffaker, Dominique A. Kagele, Marah C. Runtsch, Erin Bake, Aadel A. Chaudhuri, June L. Round and Ryan M. O'Connell J Immunol Submit online. Every submission reviewed by practicing scientists ? is published twice each month by http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://www.jimmunol.org/content/suppl/2013/05/17/jimmunol.130035 1.DC1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 26, 2021. Published May 17, 2013, doi:10.4049/jimmunol.1300351 The Journal of Immunology MicroRNA-155 Confers Encephalogenic Potential to Th17 Cells by Promoting Effector Gene Expression Ruozhen Hu,* Thomas B. Huffaker,* Dominique A. Kagele,* Marah C. Runtsch,* Erin Bake,* Aadel A. Chaudhuri,† June L.
    [Show full text]
  • Original Article Unbalanced Upregulation of Ryanodine Receptor 2 Plays a Particular Role in Early Development of Daunorubicin Cardiomyopathy
    Am J Transl Res 2015;7(7):1280-1294 www.ajtr.org /ISSN:1943-8141/AJTR0010909 Original Article Unbalanced upregulation of ryanodine receptor 2 plays a particular role in early development of daunorubicin cardiomyopathy Dana Kucerova1,2, Gabriel Doka1, Peter Kruzliak3, Katarina Turcekova1, Jana Kmecova1, Zuzana Brnoliakova4, Jan Kyselovic1, Uwe Kirchhefer2, Frank U Müller2, Peter Krenek1, Peter Boknik2, Jan Klimas1 1Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovak Re- public; 2Institut für Pharmakologie und Toxikologie, Universitätsklinikum Münster, Münster, Germany; 3Internation- al Clinical Research Center, St. Anne’s University Hospital and Masaryk University, Brno, Czech Republic; 4Institute of Experimental Pharmacology, Slovak Academy of Sciences, Bratislava, Slovak Republic Received May 31, 2015; Accepted July 14, 2015; Epub July 15, 2015; Published July 30, 2015 Abstract: Calcium release channel on the sarcoplasmic reticulum of cardiomyocytes (ryanodine receptor type 2, RyR2) plays a critical role in the regulation of calcium and was identified as a crucial factor for development of chronic anthracycline cardiomyopathy. Its early stages are less well described although these determine the later development. Hence, we tested the effect of repeated, short-term anthracycline (daunorubicin) administration on cardiac performance, cardiomyocyte function and accompanied changes in calcium regulating proteins expression. Ten-twelve weeks old male Wistar rats were administered with 6 doses of daunorubicin (DAU, 3 mg/kg, i.p., every 48 h), controls (CON) received vehicle. Left ventricular function (left ventricular pressure, LVP; rate of pressure de- velopment, +dP/dt and decline, -dP/dt) was measured using left ventricular catheterization under tribromethanol anaesthesia (15 ml/kg b.w.).
    [Show full text]
  • 1 Novel Expression Signatures Identified by Transcriptional Analysis
    ARD Online First, published on October 7, 2009 as 10.1136/ard.2009.108043 Ann Rheum Dis: first published as 10.1136/ard.2009.108043 on 7 October 2009. Downloaded from Novel expression signatures identified by transcriptional analysis of separated leukocyte subsets in SLE and vasculitis 1Paul A Lyons, 1Eoin F McKinney, 1Tim F Rayner, 1Alexander Hatton, 1Hayley B Woffendin, 1Maria Koukoulaki, 2Thomas C Freeman, 1David RW Jayne, 1Afzal N Chaudhry, and 1Kenneth GC Smith. 1Cambridge Institute for Medical Research and Department of Medicine, Addenbrooke’s Hospital, Hills Road, Cambridge, CB2 0XY, UK 2Roslin Institute, University of Edinburgh, Roslin, Midlothian, EH25 9PS, UK Correspondence should be addressed to Dr Paul Lyons or Prof Kenneth Smith, Department of Medicine, Cambridge Institute for Medical Research, Addenbrooke’s Hospital, Hills Road, Cambridge, CB2 0XY, UK. Telephone: +44 1223 762642, Fax: +44 1223 762640, E-mail: [email protected] or [email protected] Key words: Gene expression, autoimmune disease, SLE, vasculitis Word count: 2,906 The Corresponding Author has the right to grant on behalf of all authors and does grant on behalf of all authors, an exclusive licence (or non-exclusive for government employees) on a worldwide basis to the BMJ Publishing Group Ltd and its Licensees to permit this article (if accepted) to be published in Annals of the Rheumatic Diseases and any other BMJPGL products to exploit all subsidiary rights, as set out in their licence (http://ard.bmj.com/ifora/licence.pdf). http://ard.bmj.com/ on September 29, 2021 by guest. Protected copyright. 1 Copyright Article author (or their employer) 2009.
    [Show full text]
  • (CELF1), an RNA Binding Protein
    ROLES OF CUG-BP, ELAV-LIKE FAMILY MEMBER 1 (CELF1), AN RNA BINDING PROTEIN, DURING VERTEBRATE HEART DEVELOPMENT by YOTAM N. BLECH-HERMONI Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy Advisor: Dr. Andrea N. Ladd Program in Cell Biology, Department of Molecular Biology and Microbiology, School of Medicine CASE WESTERN RESERVE UNIVERSITY January 2015 CASE WESTERN RESERVE UNIVERSITY SCHOOL OF GRADUATE STUDIES We hereby approve the dissertation of Yotam N. Blech-Hermoni Candidate for the degree of Doctor of Philosophy*. Committee Chair Dr. Donna M. Driscoll Committee Member Dr. Alan M. Tartakoff Committee Member Dr. Hua Lou Date of Defense November 20, 2014 *Published material is included in accordance with contracts and stipulations set forth by the publishers. i Table of Contents List of Tables ........................................................................................................................... vii List of Figures ........................................................................................................................ viii Acknowledgements ................................................................................................................. x List of Abbreviations ............................................................................................................. xi Abstract ................................................................................................................................... xiv Chapter 1: Introduction .......................................................................................................
    [Show full text]
  • Transcriptional Profiling Reveals Extraordinary Diversity Among
    TOOLS AND RESOURCES Transcriptional profiling reveals extraordinary diversity among skeletal muscle tissues Erin E Terry1, Xiping Zhang2, Christy Hoffmann1, Laura D Hughes3, Scott A Lewis1, Jiajia Li1, Matthew J Wallace1, Lance A Riley2, Collin M Douglas2, Miguel A Gutierrez-Monreal2, Nicholas F Lahens4, Ming C Gong5, Francisco Andrade5, Karyn A Esser2, Michael E Hughes1* 1Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, United States; 2Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, United States; 3Department of Integrative, Structural and Computational Biology, The Scripps Research Institute, La Jolla, United States; 4Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States; 5Department of Physiology, University of Kentucky School of Medicine, Lexington, United States Abstract Skeletal muscle comprises a family of diverse tissues with highly specialized functions. Many acquired diseases, including HIV and COPD, affect specific muscles while sparing others. Even monogenic muscular dystrophies selectively affect certain muscle groups. These observations suggest that factors intrinsic to muscle tissues influence their resistance to disease. Nevertheless, most studies have not addressed transcriptional diversity among skeletal muscles. Here we use RNAseq to profile mRNA expression in skeletal, smooth, and cardiac muscle tissues from mice and rats. Our data set, MuscleDB, reveals extensive transcriptional diversity, with greater than 50% of transcripts differentially expressed among skeletal muscle tissues. We detect mRNA expression of *For correspondence: hundreds of putative myokines that may underlie the endocrine functions of skeletal muscle. We [email protected] identify candidate genes that may drive tissue specialization, including Smarca4, Vegfa, and Competing interests: The Myostatin.
    [Show full text]
  • Cardiac Atrial Compartmentalisation Proteomics: a Modified Density 2 Gradient Method to Analyse Endo-Lysosomal Proteins
    bioRxiv preprint doi: https://doi.org/10.1101/2021.02.22.432193; this version posted February 22, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Cardiac Atrial Compartmentalisation Proteomics: A Modified Density 2 Gradient Method to Analyse Endo-lysosomal Proteins 3 Thamali Ayagama1*, Samuel J Bose1*, Rebecca A Capel1*, David A Priestman1, Georgina 4 Berridge2, Roman Fisher2, Antony Galione1, Frances M Platt1, Holger Kramer3, Rebecca A B 5 Burton1# 6 7 1. University of Oxford, Department of Pharmacology, OX1 3QT Oxford, United Kingdom 8 2. Target Discovery Institute, University of Oxford, OX3 7FZ Oxford, United Kingdom 9 3. Biological Mass Spectrometry and Proteomics Facility, MRC London Institute of Medical 10 Sciences, Imperial College London, W12 0NN London, United Kingdom. 11 *Joint first authors; #corresponding author 12 Lead contact: [email protected] 13 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.02.22.432193; this version posted February 22, 2021. 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. 14 Summary 15 16 The importance of lysosomes in cardiac physiology and pathology are well 17 established, and evidence for roles in calcium signalling are emerging. We describe a 18 label-free proteomics method suitable for small cardiac tissue biopsies based on 19 density-separated fractionation, which allows study of endo-lysosomal (EL) proteins. 20 21 Density gradient fractions corresponding to tissue lysate; sarcoplasmic reticulum (SR), 22 mitochondria (Mito) (1.3 g/ml ); and EL with negligible contamination from SR or Mito 23 (1.04 g/ml), were analysed using Western Blot, enzyme activity assay and LC-MS/MS 24 analysis (adapted discontinuous Percoll, and sucrose differential density gradient).
    [Show full text]
  • Supplemental Table S-2. Genes That Were Differentially Regulated by 2-Fold Or Greater Between Male and Female Tendons
    Supplemental Table S-2. Genes that were differentially regulated by 2-fold or greater between male and female tendons. Log Fold Affymetrix Change Male vs Probe ID Entrez ID Symbol Description Female 17543785 213742 Xist inactive X specific transcripts -5.47 17426022 100041687 Mup8 major urinary protein 8 -3.73 17426062 100048885 Gm2083 major urinary protein LOC100048885 -3.53 17426043 100048885 Gm2083 major urinary protein LOC100048885 -3.50 17297591 NA NA NA -3.50 17426011 100048885 Gm2083 major urinary protein LOC100048885 -3.46 17425990 100048885 Gm2083 major urinary protein LOC100048885 -3.46 17425915 100048885 Gm2083 major urinary protein LOC100048885 -3.46 17425941 100048885 Gm2083 major urinary protein LOC100048885 -3.40 17517360 66402 Sln sarcolipin -3.18 17426032 100048885 Gm2083 major urinary protein LOC100048885 -3.12 17346069 240120 Zfp119b zinc finger protein 119b -3.07 17426000 100048885 Gm2083 major urinary protein LOC100048885 -3.03 17425900 100048885 Gm2083 major urinary protein LOC100048885 -2.91 17425888 100048885 Gm2083 major urinary protein LOC100048885 -2.85 17315002 70152 Mettl7a1 methyltransferase like 7A1 -2.62 17248348 NA NA NA -2.41 17524209 NA NA NA -2.39 17443890 24135 Zfp68 zinc finger protein 68 -2.38 17238834 NA NA NA -2.33 17343251 108121 U2af1 U2 small nuclear ribonucleoprotein auxiliary factor (U2AF) 1 -2.32 17406421 99889 Arfip1 ADP-ribosylation factor interacting protein 1 -2.30 17497612 93747 Echs1 enoyl Coenzyme A hydratase, short chain, 1, mitochondrial -2.22 17238892 NA NA NA -2.22 17238934 64009
    [Show full text]