Generation of a Synthetic Transcription Activator Protein Targeting the Human Fetal Γ-Globin Gene

Total Page:16

File Type:pdf, Size:1020Kb

Generation of a Synthetic Transcription Activator Protein Targeting the Human Fetal Γ-Globin Gene Generation of a synthetic transcription activator protein targeting the human fetal γ-globin gene By Maclean Bassett An honors thesis presented to the college of Liberal Arts and Sciences University of Florida 2019 0 Table of Contents 1. Abstract – Page 2 2. Introduction – Page 3 3. Materials and Methods – Page 8 4. Results – Page 12 5. Discussion and Perspective – Page 15 6. Figures – Page 17 7. Acknowledgements – Page 29 8. Abbreviations – Page 30 9. References – Page 31 1 1. Abstract: The human β-globin gene locus is located on chromosome 11 and constitutes five β-type globin genes that are organized in a manner reflecting their expression during development: an embryonic ɛ-globin gene located at the 5’ end, followed by the two fetal γ-globin genes, and the adult β- and δ- globin genes at the 3’ end. Elevated expression of fetal hemoglobin in the adult ameliorates the symptoms associated with β- hemoglobinopathies like sickle cell disease. A newly discovered DNase I hypersensitive site (HS) located 4 kb upstream of the γ-globin gene (HBG-4kb HS) was targeted as a candidate for affecting expression of γ-globin. Previous work conducted by our research group demonstrated that introduction of a synthetic zinc finger DNA-binding domain (ZF-DBD) to this site blocked recruitment of transcription factors, thus affecting expression of γ-globin. Considering this, a 6 ZF ZF-DBD was generated with a VP64 activator domain joined to the C terminus. The VP64 domain consists of 4 VP16 domains each of which encode viral proteins involved in activation of transcription by recruitment of co-activators. The synthetic protein was shown to interact with the target DNA sequence. Direct delivery of the generated ZF-DBD to mouse erythroleukemia cells revealed that the presence of the VP64 activation domain may not impede direct protein delivery to erythroid cells. 2 2. Introduction: Hemoglobin Hemoglobin is the basic protein component of human red blood cells, and functions as a respiratory carrier for transport of oxygen from the lungs to the tissues and return of carbon dioxide from the tissues to the lungs. Adult human hemoglobin possesses a heterotetrametric quaternary structure composed of four globular subunits: two α subunits and two β subunits. Each globin subunit consists of eight alpha helices which form a unique ‘globin fold’ and a non-protein heme group. Each heme group is bound to a gap between the helices and is composed of an Iron cation complexed in a porphyrin plane that forms four stable Fe-N bonds (Fig. 2.1). It is attached to the globin subunit via the central iron coordinating to the nitrogen(τ) of a proximal histidine residue found on one of the alpha helices.1 When the hemoglobin is oxygenated in the lung the oxygen becomes bound to the vacant sixth coordination position. When the Heme group is deoxygenated, the porphyrin ring sits in a nonplanar orientation to the iron atom, which is pulled toward the histidine residue. When an oxygen molecule binds, the porphyrin ring adopts a configuration planar with the iron atom, which draws the histidine residue towards the ring complex. This change in conformation induces a cooperativity effect, whereby the alpha helix becomes shifted, which alters the shape of congruent alpha helices that allows higher affinity for oxygen binding.2 Human expression of hemoglobin follows a specific timing of subunit expression constituted by a series of hemoglobin switching events. The first eight weeks of human development constitute the embryonic stage, during which embryonic hemoglobin (HbE) is composed of two ζ subunits and two ε subunits. Following embryonic development fetal development occurs. Fetal hemoglobin (HbF) is constituted of two α subunits and two γ subunits. HbF is the primary form of hemoglobin for approximately six months until shortly after birth when a second hemoglobin switching event occurs. Hemoglobin switching is characterized by a marked decrease in γ subunit production and activation of β globin subunits. The adult hemoglobin (HbA) is now composed of two α subunits and two β subunits (Fig. 2.1), which will remain the primary form of hemoglobin throughout adult life.3 This shift in hemoglobin expression is dictated by the physical proximity of the globin genes on chromosome 11, whereby their placement order follows their expression according to the general structure of 5’ ---- 3’. Control of the timing of gene activation is done by gene proximal regulatory DNA elements and the Locus Control Region (LCR). The LCR is a cis regulatory element composed of five DNAse I Hypersensitive Sites (HS). These HS elements contain multiple transcription factor motifs. The HS elements work cooperatively at the globin gene loci to mediate high levels of globin expression in time with developmental stages.3 The actual mechanism of 3 the switch is in part mediated by competition for the LCR by the and gene loci4, but primarily by autonomous silencing of the -globin gene locus.5 Hemoglobinopathies and Hereditary Persistence of Fetal Hemoglobin (HPFH) Diseases that affect the synthesis of functional hemoglobin are extremely common globally, there are more than 1,000 naturally occurring hemoglobin mutants that occur in humans6 which alter structure and biochemical properties of the hemoglobin. Six hundred diseases have hence been defined by the American College of Medical Genetics, notably including Sickle Cell Anemia and Beta Thalassemia, which affect the ability of hemoglobin to carry oxygen effectively.7 These diseases are congenital, as hemoglobinopathies are linked to genetic heritage, most notoriously Sickle Cell Anemia resulting from the inheritance of an abnormal hemoglobin S gene from both parents. An estimated 7% of the world’s population is affected by a form of hemoglobin mutation.8 In rare instances, usually due to a point mutation in the γ-globin gene promoter or deletions in the -gene loci9, HbF production continues into adulthood. This condition, known as Hereditary Persistence of Fetal Hemoglobin (HPFH), is harmless to the carrier and is characterized by elevated levels of HbF in the blood in addition to HbA. Increased levels of HbF in the blood have been shown to ameliorate complications associated with β-globin disorders10, especially so in the case of Sickle Cell Anemia where high levels of HbF were demonstrated to prevent basic mechanisms that lead to the development of Sickle Cell Anemia. Sickle cell anemia is the pathological culmination of conditions caused by the polymerization of a mutant form of -hemoglobin subunits named Hemoglobin S (HbS). Hemoglobin S subunits polymerize together upon deoxygenation, causing the cell’s shape to distort into that iconic sickle shape. HbF directly disrupts the polymerization of HbS.11 Furthermore, it decreases the relative concentration of HbS in the blood and is capable of binding oxygen more tightly than HbA, both of which effectively compensates for the defective HbS.12 In patients where HbS makes up over 50% of their total hemoglobin, clinical Sickle Cell Anemia is observed, however when blood hemoglobin is composed of at least 10% the severity of Sickle Cell Anemia is highly diminished.13 The occurrence of HPFH is genetically linked and there is a clinical pedigree whereby an individual inherits HPFH from one parent and Sickle Cell Anemia from the other parent (HbS/HPFH) which leads to a concentration of HbF in the blood well above this threshold. Although, increasing levels of HbF in the blood is by no means a cure, and even with heightened levels of HbF life threatening complications can still arise in patients.14 However, finding methods of increasing HbF concentration levels in 4 the blood represents the most promising strategy for developing treatments for Sickle Cell Anemia due to its ability to impact the physiological symptoms of the disease. Fetal globin gene silencing and reactivation The mechanisms that modulate the silencing of the -globin genes during hemoglobin switching have remained largely elusive, however efforts to understand the proteins and pathways that comprise the “big picture” have shed light on the role of BCL11A in - globin gene silencing. The BCL11A gene, located on chromosome 2, is a highly conserved gene sequence containing multiple Single Nucleotide Polymorphisms (SNPs) associated with elevated fetal hemoglobin expression. BCL11A contains three C2H2-type zinc finger (ZF) motifs, a proline rich region, and an acidic domain. The BCL11A zinc fingers bind directly to GG-rich sequences and BCL11A functions as transcriptional repressor.15 The SNPs are highly associated with F-cell levels and the expression of HbF16 as well as prediction of pain crises in sickle cell disease.17 BCL11A acts within the β-globin locus to control the silencing of -globin gene18, however the exact mechanism that modulates this silencing is unknown. Mutations in the BCL11A gene have thus been associated with HPFH.19 20 Furthermore, the silencing of an enhancer associated GATA motif present in the BCL11A locus results in elevated expression of HbF.21 Synthetic transcription activators Synthetic transcription activators or artificial transcription factors are proteins developed to target and modulate the transcription of specific genes in the genome. Coupling a transcription activation domain with a zinc finger DNA-binding domain (DBD) allows for targeted delivery of the transcription activator to a target site. In the case of an activator domain, delivery by ZF-DBDs results in the upregulation of the target gene product.22 VP16 is a transactivation domain found in the Herpes simplex 1, virus which primes transcription of the virally encoded IE genes. VP16 contains a carboxy-terminal region with an acidic sequence at its terminus that is crucial to gene activation.23 This terminal motif can be attached to DNA-binding domains that recognize specific promotor sequences to activate transcription.24 Hence, linkage of a ZF-DBD to a VP16 domain achieves activation of specific target genes.25 5 Zinc Fingers (ZFs) are protein/DNA interaction domains capable of targeting specific sequences of DNA.
Recommended publications
  • Fetal and Embryonic Haemoglobins P
    Review Article J Med Genet: first published as 10.1136/jmg.10.1.50 on 1 March 1973. Downloaded from Journal of Medical Genetics (1973). 10, 50. Fetal and Embryonic Haemoglobins P. A. LORKIN MRC Abnormal Haemoglobin Unit, University Department of Biochemistry, Cambridge Haemoglobin has been the subject of intensive form a nearly spherical molecule with extensive research for many years and is one of the most areas of contact between unlike chains; the two thoroughly understood of all protein molecules. main types of contact are denoted alp, and alg2 The amino-acid sequences of haemoglobins from The tetramer exhibits cooperative behaviour or many species of animals have been determined haem-haem interaction. As each haem combines (tabulated by Dayhoff, 1969) and the molecular with oxygen the affinity of successive haems in- structures of horse and human haemoglobins have creases. The oxygen affinity curve of the tetramer been determined in great detail by x-ray crystallo- is sigmoidal and may be represented approximately graphy (Perutz et al, 1968a and b; Perutz 1969). A by the Hill equation:* mechanism of action of haemoglobin has been pro- = kpo2n posed (Perutz, 1970a and b and 1972). The y haemoglobins of higher organisms share a common +kpo2n tetrameric structure built up of two pairs of unlike Oxygen affinity data are usually presented in copyright. chains; the a chains containing 141 amino-acid terms of P102, the partial pressure of oxygen re- residues and the non-a chains containing generally quired to attain half saturation with oxygen, and of 145 or 146 amino acids. In man, five types of n, the exponent of the Hill equation.
    [Show full text]
  • Linkage of Genes for Adult A-Globin and Embryonic Aulike Globin Chains (Hemoglobin H/Thalassemia/Embryonic Hemoglobin/Mice) J
    Proc. Natl. Acad. Sci. USA Vol. 77, No. 2, pp. 1087-1090, February 1980 Genetics Linkage of genes for adult a-globin and embryonic aulike globin chains (hemoglobin H/thalassemia/embryonic hemoglobin/mice) J. BARRY WHITNEY III* AND ELIZABETH S. RUSSELL The Jackson Laboratory, Bar Harbor, Maine 04609 contributed by Elizabeth S. Russell, November 19, 1979 ABSTRACT In a-thalassemia, the genetic locus for the a (Hbaa) female mouse and a triethylene-melamine-treated male chains of adult hemoglobin is not expressed. We have examined that carried a different, doublet, Hba haplotype (5). In this the hemoglobins of a number of individual mouse embryos affected a-thalassemic offspring of the treated male, only the heterozygous for a particular a-thalassemia (Hba th-J) and find n'o.'ecrease in the proportion of hemoglobins containing the Hbaa haplotype inherited from the C57BL/6J mother was a chain as compared to the hemoglobin containing the a-like expressed. The hemoglobins of this mouse were found by embryonic globin chain. This result suggests that the locus for electrophoresis after cystamine treatment (6) to be unusual in this embryonic a-like chain is inactivated or deleted in these that they contained a lower than normal proportion of the he- embryos as well. Because a single mutational event inactivated moglobin containing the diffuse-major:/ chain, a condition also adult and embryonic loci, we conclude that they are probably reported for the x-ray-induced mouse a-thalassemia discovered closely linked to one another on the same chromosome. We also present evidence that an unusual hemoglobin in the blood of at the Oak Ridge National Laboratory (7).
    [Show full text]
  • WO 2017/070364 Al 27 April 2017 (27.04.2017) P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2017/070364 Al 27 April 2017 (27.04.2017) P O P C T (51) International Patent Classification: AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, A61K 39/395 (2006.01) C07K 16/18 (2006.01) BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, C07K 16/00 (2006.01) DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KN, KP, KR, (21) International Application Number: KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, PCT/US20 16/057942 MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, (22) International Filing Date: OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, 20 October 2016 (20.10.201 6) SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, (25) Filing Language: English ZW. (26) Publication Language: English (84) Designated States (unless otherwise indicated, for every (30) Priority Data: kind of regional protection available): ARIPO (BW, GH, 62/244,655 2 1 October 2015 (21. 10.2015) US GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, (71) Applicant: QOOLABS, INC. [US/US]; 4186 Sorrento TJ, TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, Valley Blvd., Suite D/E, San Diego, CA 92121 (US).
    [Show full text]
  • Sourcebook in Forensic Serology, Immunology, and Biochemistry
    UNIT VII. HEMOGLOBIN, SERUM GROUP SYSTEMS, HLA AND OTHER GENETIC MARKERS Hemoglobin SECTION 38. HEMOGLOBIN 38.1 Introduction Hemoglobin (Hb)is the major protein of human red cells, forth. Hemoglobins exist which contain only one kind of comprising about 95% of their dry weight. Adult human chain: Hb H is B4, for example, and Hb Bart's is 7,. blood normally contains from about 4 million to 6.5 million Jones (1961) pointed out that hemoglobin structural red cells per mm3 blood, the average figure being slightly heterogeneity can be classified as follows: (1) Maturation higher for men. Hemoglobin itself is present in concentra- heterogeneity, which refers to the fact that different tions of about 14 to 16 g per 100 mP blood. It is the oxygen hemoglobins are normally synthesized during different transporting protein in higher animals; without a molecule stages of development. There are embryonic, fetal and adult having its properties, complex multicellular aerobic life as hemoglobins. (2) Minor hemoglobin heterogeneity, which we know it would not be possible. refers to the presence of small amounts of structurally dif- Hemoglobin is one of the most extensively studied of all ferent but normal hemoglobins along with the major proteins, and its literature fills many volumes. As noted in component characteristic of a particular stage of develop- section 5.1, it acquired its present name over 100 years ago ment; and (3) genetic heterogeneity, which refers to the (Hoppe-Seyler, 1864). In forensic serology, hemoglobiin is various "abnormal" hemoglobin variants. Many of these important in two principal contexts: (1) Blood is normally are thought to be the result of point mutations, and with a idenad in questioned samples by procedures designed to few exceptions, variant hemoglobins are very rare.
    [Show full text]
  • The Free Alpha-Hemoglobin: a Promising Biomarker for Β-Thalassemia
    r Biomar ula ke c rs le o & M D f i a o g l Journal of Molecular Biomarkers n a o n Abdullah, et al., J Mol Biomark Diagn 2014, 5:5 r s i u s o J DOI: 10.4172/2155-9929.1000197 ISSN: 2155-9929 & Diagnosis Review Article Open Access The Free Alpha-Hemoglobin: A Promising Biomarker for β-Thalassemia Uday YH Abdullah1, Ahmed GF Al-Attraqchi1, Hishamshah M Ibrahim2, Zilfalil Bin Alwi3, Atif A Baig1, Iekhsan Othman4, Noraesah B Mahmud2, Rosline B Hassan3, Nor Hedayah A Bakar1 and Alawiyah B A Abd Rahman5 1University Sultan Zainal Abidin (UniSZA), Faculty of Medicine and Health Sciences, Kuala Terengganu, Malaysia 2Hospital Kuala Lumpur (HKL), Kuala Lumpur, Malaysia 3Univesiti Sains Malaysia (USM), School of Medical Science, Kelantan, Malaysia 4Monash University, Selangor, Malaysia 5Hospital Sultana Nor Zahirah (HSNZ), Kuala Terengganu, Malaysia *Corresponding author: Uday Younis Hussein Abdullah, University Sultan Zainal Abidin (UniSZA), Kampus Kota, Jalan Sultan Mahmud 20400, Kuala Terengganu, Malaysia, Tel: 609-625607; Fax: 60-6275771/5772; E-mail: [email protected] Rec date: Aug 28, 2014; Acc date: Sep 27, 2014; Pub date: Sep 29, 2014 Copyright: © 2014 Abdullah UYH, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Abstract The purpose is to review the current knowledge and methods of determination of the free alpha hemoglobin with emphasis on its potential implications in patients with beta-thalassemia (β-thal).The severity of β-thal correlates with the extent of imbalance between α- and non-α-globin chains and the amount of the free alpha hemoglobin (free α- Hb) pool in the erythrocytes.
    [Show full text]
  • BET Inhibitors Enhance Embryonic and Fetal Globin Expression in Erythroleukemia Cell Lines by John Z
    BET inhibitors enhance embryonic and fetal globin expression in erythroleukemia cell lines by John Z. Cao, Kristina Bigelow, Amittha Wickrema, and Lucy A. Godley Received: March 16, 2021. Accepted: August 20, 2021. Citation: John Z. Cao, Kristina Bigelow, Amittha Wickrema, and Lucy A. Godley. BET inhibitors enhance embryonic and fetal globin expression in erythroleukemia cell lines. Haematologica. 2021 Aug 26. doi: 10.3324/haematol.2021.278791. [Epub ahead of print] Publisher's Disclaimer. E-publishing ahead of print is increasingly important for the rapid dissemination of science. Haematologica is, therefore, E-publishing PDF files of an early version of manuscripts that have completed a regular peer review and have been accepted for publication. E-publishing of this PDF file has been approved by the authors. After having E-published Ahead of Print, manuscripts will then undergo technical and English editing, typesetting, proof correction and be presented for the authors' final approval; the final version of the manuscript will then appear in a regular issue of the journal. All legal disclaimers that apply to the journal also pertain to this production process. BET inhibitors enhance embryonic and fetal globin expression in erythroleukemia cell lines John Z. Cao1, Kristina Bigelow2, Amittha Wickrema1,2, and Lucy A. Godley1,2* 1 Committee on Cancer Biology, Biological Sciences Division, The University of Chicago, Chicago IL 2 Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago IL *Corresponding author Contact information: 5841 S. Maryland Ave. MC 2115 Chicago, IL 60637 Phone: 773-702-4140 FAX: 773-702-9268 Email: [email protected] Author Contribution J.Z.C.
    [Show full text]
  • Fetal Hemoglobin Induction to Treat Β-Hemoglobinopathies
    Central Journal of Hematology & Transfusion Review Article Corresponding author Betty Pace, Department of Pediatrics, Georgia Regents University, 1120 15th St. CN-4112, Augusta, GA 30912, Fetal Hemoglobin Induction to USA, Tel: (706) 721-6893; Email: Submitted: 21 May 2014 Treat b-Hemoglobinopathies: Accepted: 01 June 2014 Published: 05 June 2014 ISSN: 2333-6684 From Bench to Bedside Copyright Makala LH1,2, Torres CM1, Clay EL1,2,3, Neunert C1,2 and Betty S. © 2014 Pace et al. Pace1,2,4* OPEN ACCESS 1Department of Pediatrics, Georgia Regents University, US 2Cancer Center Member, Georgia Regents University, US Keywords 3Department of Internal Medicine, Georgia Regents University, US • Fetal hemoglobin 4Biochemistry and Molecular Biology, Georgia Regents University, US • γ-globin • Sickle cell disease • Hydroxyurea Abstract The developmental regulation of the - to -globin switch has motivated research ef- forts to establish therapeutic modalities for individuals affected with -hemoglobinopathies. Fetal Hemoglobin (HbF) synthesis is high atγ birthβ but declines to adult levels by one year of ages; since HbF blocks hemoglobin S polymerization in Sickle Cellβ Disease (SCD) and compensated for anemia in -Thalassemia, reactivating expression is of great research interest. Naturally occurring mutations in the -globin locus on chromosome 11, produce elevated HbF expression afterβ birth which ameliorates symptoms in SCD. Genome-wide studies discovered three gene loci includingβ BCL11A that account for the majority of inherited HbF variance but considerable work is required to advance this protein as a therapeutic target. Therefore, efforts to develop chemical inducers of HbF are desirable. Hydroxyurea is a potent HbF inducer with clinical efficacy in adults and children with SCD but it has been underutilized in the clinical setting.
    [Show full text]
  • California State University, Northridge Gene
    CALIFORNIA STATE UNIVERSITY, NORTHRIDGE GENE EXPRESSION PROFILING IN A FAMILY WITH A NOVEL FORM OF BETA-THALASSEMIA A thesis submitted in partial fulfillment of the requirement For the degree of Master of Science in Biology By Forough Taghavifar May 2016 The thesis of Forough Taghavifar is approved: Dr. Stan Metzenberg, Ph.D. Date Dr. Steven B Oppenheimer, Ph.D. Date Dr. Aida Metzenberg, Ph.D., Chair Date California State University, Northridge ii ACKNOWLEDGEMENTS I would like to extend the outmost gratitude to my father, mother and husband whose emotional support and guidance has enabled me to successfully enhance my academic journey. Also, a heartfelt appreciation goes to Dr. Aida Metzenberg, my wise and trusting advisor. Dr. Aida Metzenberg, not only have you been my thesis advisor and mentor, you have also served as a mother figure for me ever since I emigrated to the US and started my master’s program at Cal State University Northridge, -for that I will always thank you! Furthermore, I would thank Dr. Stan Metzenberg for believing in me before I personally believed in my abilities. Dr. Stan Metzenberg, if it were not for your computer modeling class, a spark would not have ignited my interest in wanting to further sophisticate my project. Lastly, I would like to acknowledge the Bruins in Genomics (BIG) summer program that I had the pleasure of attending in the summer of 2015. Also a very special thanks to my mentor at the BIG program, David Casero, who has more than willingly assisted me throughout all the RNA sequencing data analysis steps.
    [Show full text]
  • Hoppe-Seyler's Zeitschrift Für Physiologische Chemie
    Hoppe-Seyler's Zeitschrift für Physiologische Chemie Band 365 — 2. Jahreshälfte Fortgeführt von A. Kossei, F.Knoop, K.Thomas, F. Lynen, A. Butenandt und G. Weitzel Herausgegeben von K.Decker, W. Stoffel, H.G.Zachau unter Mitwirkung von H. Eggerer, J. Engel, H. Fritz, E. Habermann, E. Helmreich, A. Henschen, B. Hess, N. Hilschmann, H. Hilz, P.W. Jungblut, K. Jungermann, P. Karlson, H. Kleinkauf, H.L. Kornberg, K. Kühn, D. Oesterhelt, K. Rajewsky, K. Sandhoff, R. Schauer, J. Seelig, G. Siebert, H. Sies, H. Simon, E. Truscheit, H. Tschesche, H. Tuppy, V. Ullrich, E. Wecker, H. Wiegandt, Β. Wittmann-Liebold, H. Zahn Redaktion A. Dillmann, G. Peters W DE G 1984 WALTER DE GRUYTER · BERLIN · NEW YORK Gesamtregister 1984 Stichwortregister yV-Acetyl-9-Ö-acetylneuraminic acid, in terminal posi• Adipocytes, (rat) insulin receptor complex, G. Klotz, tion in a disialoganglioside (Grj)la), D.C.Gowda, D.J. Saunders and D. Brandenburg, G. Reuter, A.K. Shukla and R. Schauer, 1247 493 2-Acetyl- 1-0-alkylglycero-phosphocholines, platelet Adipokinetic hormones, purification and amino-acid aggregation and cAMP concentration, I. Η ill mar, composition, G. Gäde, G.J. Goldsworthy, Τ. Muramatsu and Ν. Zöllner, 33 G.Kegel and R.Keller, 393 Acetylcholinesterase (EC 3.1.1.7), (Electrophorus - (of Manduca sexta), amino-acid composition, electricus), fluorescence-labelled inhibitors, R. Ziegler, G. Kegel and R. Keller, 1451 W. Hallenbach and L. Horner, 1475 ADP-Ribosyltransferase (EC 2.4.-.—), Ascaris suum, O-Acetyl-Grjia, s. TV-acetyl-9-O-acetylneuraminic acid R.D.Walter and E. Ossikovski, 805 /3-JV-Acetyl-D-hexosaminidase (EC 3.2.1.52), (in cultured Affinity chromatography, rat liver lysosomes, F.-J.
    [Show full text]
  • Homozygous Α-Thalassemia in a Growth Retarded, Non-Hydropic
    Journal of Perinatology (2008) 28, 158–159 r 2008 Nature Publishing Group All rights reserved. 0743-8346/08 $30 www.nature.com/jp PERINATAL/NEONATAL CASE PRESENTATION Homozygous a-thalassemia in a growth retarded, non-hydropic premature newborn YPM Ng1, R Joseph1,2 and A Biswas3,4 1Department of Neonatology, National University Hospital, Singapore; 2Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; 3Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore and 4Department of Obstetrics and Gynecology, National University Hospital, Singapore infusions, 20 ml kgÀ1 normal saline, repeated infusions of 4.2% A newborn with homozygous a-thalassemia presented with intrauterine sodium bicarbonate and empiric antibiotics. growth retardation and presumed persistent pulmonary hypertension. The baby’s hemoglobin at 2 h of life was 11.8 g per 100 ml. The He also had moderate anemia, hepatomegaly and hypospadias. Correlating peripheral blood film showed anisopoikilocytosis with numerous the newborn’s clinical presentation with an underlying cause of anemia was nucleated red blood cells (1489 per 100 white blood cells), target cells helpful for early diagnosis. Prenatal blood tests must include red cell indices and Howell Jolly bodies. The cause of anemia1 and low oxygen carrying and a mean corpuscular volume value below 80 fL should prompt capacity of the blood was suspected to be a severe hemoglobinopathy thalassemia screening in an at-risk population. although his parents were not known thalassemia carriers. Journal of Perinatology (2008) 28, 158–159; doi:10.1038/sj.jp.7211873 At 5 h of life, a transfusion of 10 ml kgÀ1 packed red cells Keywords: anemia; Bart’s hydrops; homozygous a-thalassemia; improved the partial pressure of oxygen and the metabolic acidosis.
    [Show full text]
  • Flipping the Hemoglobin Switch and Discovering Regulators Involved in Fetal Hemoglobin Reactivation
    Flipping the Hemoglobin Switch and Discovering Regulators Involved in Fetal Hemoglobin Reactivation By Mandy Y Boontanrart A dissertation submitted in partial satisfaction of the requirements for the degree of Doctor of Philosophy in Molecular and Cell Biology in the Graduate Division of the University of California, Berkeley Committee in charge: Professor Jacob Corn, Chair Professor Ellen Robey Professor Nicholas Ingolia Professor John Dueber Summer 2020 Abstract Flipping the Hemoglobin Switch and Discovering Regulators Involved in Fetal Hemoglobin Reactivation By Mandy Y Boontanrart Doctor of Philosophy in Molecular and Cell Biology University of California, Berkeley Professor Jacob Corn, Chair The fetal to adult hemoglobin switch is a developmental process by which fetal hemoglobin becomes silenced after birth and replaced by adult hemoglobin. Diseases caused by defective or missing adult hemoglobin, such as Sickle Cell Disease or β- Thalassemia, can be ameliorated by reactivating fetal hemoglobin. We discovered that knockdown or knockout of β-globin, a subunit of adult hemoglobin, led to robust upregulation of γ-globin, a subunit of fetal hemoglobin. This phenomenon suggested that red blood cells have an inherent ability to upregulate fetal hemoglobin in the event that adult hemoglobin is lacking. We developed multiple gene-editing tools in an immortalized erythroid cell model to investigate the molecular mechanisms behind the increase in fetal hemoglobin. Time- course transcriptomics identified ATF4, a transcription factor, as a causal regulator of this response. Further analysis also converged upon downregulation of MYB and BCL11A, known repressors of γ-globin, described in detail in chapter 2. Further work in chapter 3 explores other possible fetal hemoglobin regulators as discovered by CRISPRi arrayed mediated knockdown experiments.
    [Show full text]
  • Micromethodology for the Characterization of Hemoglobin Variants
    Micromethodology for the characterization of hemoglobin variants Citation for published version (APA): Plaseska, D. (1994). Micromethodology for the characterization of hemoglobin variants. Rijksuniversiteit Limburg. https://doi.org/10.26481/dis.19940428dp Document status and date: Published: 01/01/1994 DOI: 10.26481/dis.19940428dp Document Version: Publisher's PDF, also known as Version of record Please check the document version of this publication: • A submitted manuscript is the version of the article upon submission and before peer-review. There can be important differences between the submitted version and the official published version of record. People interested in the research are advised to contact the author for the final version of the publication, or visit the DOI to the publisher's website. • The final author version and the galley proof are versions of the publication after peer review. • The final published version features the final layout of the paper including the volume, issue and page numbers. Link to publication General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal. If the publication is distributed under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license above, please follow below link for the End User Agreement: www.umlib.nl/taverne-license Take down policy If you believe that this document breaches copyright please contact us at: [email protected] providing details and we will investigate your claim.
    [Show full text]