Adar3 Is Involved in Learning and Memory in Mice

Total Page:16

File Type:pdf, Size:1020Kb

Adar3 Is Involved in Learning and Memory in Mice ORIGINAL RESEARCH published: 13 April 2018 doi: 10.3389/fnins.2018.00243 Adar3 Is Involved in Learning and Memory in Mice Dessislava Mladenova 1,2, Guy Barry 1,2†, Lyndsey M. Konen 1,3,4, Sandy S. Pineda 1,2, Boris Guennewig 1,2†, Lotta Avesson 1, Raphael Zinn 1,3,4, Nicole Schonrock 1,2, Maina Bitar 1,2†, Nicky Jonkhout 1,2, Lauren Crumlish 5, Dominik C. Kaczorowski 1, Andrew Gong 1†, Mark Pinese 1,2, Gloria R. Franco 6, Carl R. Walkley 7, Bryce Vissel 1,3,4 and John S. Mattick 1,2* Edited by: Michael F. Miles, 1 Garvan Institute of Medical Research, Sydney, NSW, Australia, 2 St. Vincent’s Clinical School, University of New South Virginia Commonwealth University, Wales, Sydney, NSW, Australia, 3 Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of United States Technology Sydney, Sydney, NSW, Australia, 4 St. Vincent’s Centre for Applied Medical Research (AMR), Sydney, NSW, Reviewed by: Australia, 5 Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia, 6 Departamento de Sulev Kõks, Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil, University of Tartu, Estonia 7 St. Vincent’s Institute of Medical Research, Department of Medicine, St. Vincent’s Hospital, The University of Melbourne, Karine Merienne, Fitzroy, VIC, Australia Centre National de la Recherche Scientifique (CNRS), France The amount of regulatory RNA encoded in the genome and the extent of RNA editing by *Correspondence: John S. Mattick the post-transcriptional deamination of adenosine to inosine (A-I) have increased with [email protected] developmental complexity and may be an important factor in the cognitive evolution †Present Address: of animals. The newest member of the A-I editing family of ADAR proteins, the Guy Barry and Maina Bitar, vertebrate-specific ADAR3, is highly expressed in the brain, but its functional significance QIMR Berghofer Medical Research In vitro Institute, Brisbane, QLD, Australia; The is unknown. studies have suggested that ADAR3 acts as a negative regulator School of Medicine, The University of of A-I RNA editing but the scope and underlying mechanisms are also unknown. Queensland, St Lucia, QLD, Australia Meta-analysis of published data indicates that mouse Adar3 expression is highest in the Boris Guennewig, Sydney Medical School, Brain and hippocampus, thalamus, amygdala, and olfactory region. Consistent with this, we show Mind Centre, The University of that mice lacking exon 3 of Adar3 (which encodes two double stranded RNA binding Sydney, Camperdown, NSW, Australia Andrew Gong, domains) have increased levels of anxiety and deficits in hippocampus-dependent School of Medical Sciences, short- and long-term memory formation. RNA sequencing revealed a dysregulation of University of New South Wales, genes involved in synaptic function in the hippocampi of Adar3-deficient mice. We also Sydney, NSW, Australia show that ADAR3 transiently translocates from the cytoplasm to the nucleus upon Specialty section: KCl-mediated activation in SH-SY5Y cells. These results indicate that ADAR3 contributes This article was submitted to to cognitive processes in mammals. Neurogenomics, a section of the journal Keywords: ADAR3, Adar3exon3 mouse model, RNA editing, learning and memory, Adarb2 Frontiers in Neuroscience Received: 22 January 2018 Accepted: 27 March 2018 INTRODUCTION Published: 13 April 2018 Citation: The human brain has evolved to enable unique cognitive capabilities and has tripled in Mladenova D, Barry G, Konen LM, size since the split from the chimpanzee lineage around 5 million years ago. Current Pineda SS, Guennewig B, Avesson L, hypotheses suggest that the advancement of human cognition most likely arose through Zinn R, Schonrock N, Bitar M, the combined effects of the expansion of brain size and complexity, protein evolution Jonkhout N, Crumlish L, (including new splice variants) and the emergence of RNA-based regulatory mechanisms Kaczorowski DC, Gong A, Pinese M, that facilitate the epigenetic reformation of neural circuitry in response to experience Franco GR, Walkley CR, Vissel B and Mattick JS (2018) Adar3 Is Involved in (Barry and Mattick, 2012). Learning and Memory in Mice. One of the RNA-diversification/regulatory mechanisms is RNA editing. Two main types of Front. Neurosci. 12:243. RNA editing are known, one involving the deamination of cytidine to create uridine (C-to-U),and doi: 10.3389/fnins.2018.00243 the other, the deamination of adenosine to inosine (A-to-I). RNA editing not only alters the Frontiers in Neuroscience | www.frontiersin.org 1 April 2018 | Volume 12 | Article 243 Mladenova et al. ADAR3 Involved in Learning-Memory nucleotide sequence of target RNAs, but presumably also their MATERIALS AND METHODS structure-function relationships and interactions (Bass, 2002; Adar3exon3 Jantsch and Öhman, 2008). Mouse Lines: Generation of The existence of RNA editing in mammals was first discovered Mice tm1a(KOMP)Mbp in mRNAs for important neuroreceptors, such as the glutamate ES cells containing the targeted Adarb2 allele and serotonin receptors, and was initially thought to be (IKMC project number 39714; hereafter referred to as tm1a an interesting but idiosyncratic mechanism to change their Adar3 ) were generated by the trans-NIH Knock-Out amino acid sequence (Maas et al., 2003), presumably to alter Mouse Project (KOMP) and obtained from the KOMP tm1a(KOMP)Mbp the electrophysiological properties of synapses in response to Repository (www.komp.org). The Adarb2 activity or other cues. Subsequently, transcriptome-wide analyses allele contains a splice acceptor-beta-geo-polyA (SA-βgeo- revealed that A-I editing is widespread in humans, occurring pA) flanked by FRT sites located in intron 2 and loxP in thousands of transcripts, mostly in Alu sequences within elements flanking exon 3 (KOMP designation: KO first allele intronic and intergenic sequences, varying in different tissues (reporter-tagged insertion with conditional potential). Correct (Athanasiadis et al., 2004; Blow et al., 2004; Kim et al., targeting was confirmed and transgenic mice were generated 2004; Levanon et al., 2004; Ramaswami et al., 2012; Huntley from embryonic stem cell clones DEPD0006_14_A03 & et al., 2016). Although operating via a conserved mechanism DEPD0006_14_A05. Animals were generated by the Australian (Jin et al., 2009), the rates of A-to-I editing have increased Phenomics Network ES to Mouse service at Monash University. dramatically throughout vertebrate, mammalian and especially Positive mice were backcrossed to C57BL/6N background ′ primate evolution, with RNA editing in humans being more and genotyped by PCR. Primers P1 (located at the 3 end ′ ′ than an order of magnitude higher than in mouse (Kim et al., of the 5 homology arm) and P2 (located at the 5 end of 2004; Levanon et al., 2004). Moreover, more editing occurs in the exon ENSMUSE00000465454) will amplify a product of 554 human brain than in other primates (Paz-Yaacov et al., 2010). bp from the wild type allele. Primers P1 and P3 (located in Adenosine Deaminase Acting on RNA (ADAR) proteins, are the en-2 intron) will amplify a product of 246 bp from the ′ responsible for the execution of the A-to-I RNA editing through targeted allele. P1 = 5 CAATATACCACAACGAACATCTTTG ′ ′ ′ ′ hydrolytic deamination (Bass, 2002). Three ADAR enzymes 3 ; P2 = 5 GTCCCCAGGTTGCTCACATTTCG 3 ; P3 = 5 ′ (ADAR1-3) are encoded in the vertebrate genome, with ADAR3 CAACGGGTTCTTCTGTTAGTCC 3 . PCR conditions were: ◦ being vertebrate-specific (Chen et al., 2000). Common to all an initial denaturation step at 94 C 3 min, followed by 35 cycles ◦ ◦ ◦ ◦ ADARs is a C-terminal catalytic domain and multiple double- of 94 C 30s, 57 C 30s, 72 C 45s and a final cycle of 72 C for stranded RNA binding domains (Nishikura, 2010). In particular, 5 min. tm1a ADAR3 shares 50% amino-acid sequence identity with ADAR2 Heterozygous Adar3 animals (identified in-house (Melcher et al., 1996a) and is almost exclusively expressed in the as ADAR2BlacZ line) were crossed with heterozygotes nervous system, but its role is unknown. Unlike the other ADAR of a ubiquitous expressing Cre-recombinase mouse line proteins, ADAR3 contains a novel arginine rich motif (herein, R- (C57BL/6NTac-Gt(ROSA)26Sortm16(cre)Arte; Taconic). The domain), which allows the binding of single stranded RNA (Chen Cre-recombinase gene was identified using the following ′ et al., 2000), activity that may result in novel functions. Based primers, which yielded a 408 bp fragment: forward 5 ′ on in vitro evidence, ADAR3 is suggested to act as a dominant- GCATTACCGGTCGATGCAACGAGTGATGAG 3 ; and ′ negative regulator of A-to-I RNA editing (Chen et al., 2000). The reverse 5 GAGTGAACGAACCTGGTCGAAATCAGTGCG ′ R-domain has also been proposed to serve as a functional nuclear 3 . Thus, in addition to expressing Cre-recombinase, double localization signal (NLS) as it mediates interactions between heterozygous mice from the resulting cross, possessed one WT ADAR3 and the Importin α protein complex enabling ADAR3 to allele and one allele in which Cre-mediated deletion of the loxP- locate to the nucleus (Maas and Gommans, 2009) where A-to-I flanked portion of the Adar3 gene had occurred. To maintain RNA editing is believed to occur (Jin et al., 2009). the strain background and breed out the Cre-recombinase allele, Due to the novelty and relatively unknown function of double heterozygous animals were then crossed back to WT mice ADAR3 and its high expression in the nervous system, we from the ADAR2BlacZ line. Resulting heterozygous mice for the investigated the role of this protein in cognition and behavior KO and also Cre-negative, were used to maintain the colony by in ADAR3 deficient mice. We also demonstrate that mice breeding heterozygous by heterozygous. Homozygotes possessa lacking exon 3 of Adarb2 (referred to herein as Adar3exon3), non-functional ADAR3 protein and were thus referred to here as exon3 containing the double stranded RNA binding domains, display Adar3 . increased anxiety levels and impaired short and long-term hippocampus-dependent memory formation.
Recommended publications
  • A-To-I RNA Editing Does Not Change with Age in the Healthy Male Rat Brain
    Biogerontology (2013) 14:395–400 DOI 10.1007/s10522-013-9433-8 RESEARCH ARTICLE A-to-I RNA editing does not change with age in the healthy male rat brain Andrew P. Holmes • Shona H. Wood • Brian J. Merry • Joa˜o Pedro de Magalha˜es Received: 18 January 2013 / Accepted: 15 May 2013 / Published online: 26 May 2013 Ó The Author(s) 2013. This article is published with open access at Springerlink.com Abstract RNA editing is a post-transcriptional pro- Introduction cess, which results in base substitution modifications to RNA. It is an important process in generating Adenosine to inosine (A-to-I) RNA editing is a post- protein diversity through amino acid substitution and transcriptional process that alters the sequences of the modulation of splicing events. Previous studies RNA molecules. The adenosine deaminases ADAR have suggested a link between gene-specific reduc- and ADARB1 convert specific adenosine residues on tions in adenosine to inosine RNA editing and aging in RNA to inosine bases. During translation, sequencing, the human brain. Here we demonstrate that changes in and splicing, inosine is recognized as guanosine. RNA editing observed in humans with age are not Therefore, A-to-I RNA editing has important impli- observed during aging in healthy rats. Furthermore, we cations in altering specific amino acids, miRNA identify a conserved editing site in rats, in Cog3.We targeting, and in the modulation of alternative splicing propose that either age-related changes in RNA (Nishikura 2010). editing are specific to primates or humans, or that Targets of A-to-I RNA editing are often genes they are the manifestation of disease pathology.
    [Show full text]
  • Altered Regulation of Adipomir Editing with Aging
    International Journal of Molecular Sciences Article Altered Regulation of adipomiR Editing with Aging Sabel Meadows, Abbagael Seidler, Madison Wall, Jamika Page, Cara Taylor, Brendin Flinn , Robin Turner and Nalini Santanam * Department of Biomedical Sciences, Joan C Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; [email protected] (S.M.); [email protected] (A.S.); [email protected] (M.W.); [email protected] (J.P.); [email protected] (C.T.); fl[email protected] (B.F.); [email protected] (R.T.) * Correspondence: [email protected] Received: 18 August 2020; Accepted: 17 September 2020; Published: 20 September 2020 Abstract: Adipose dysfunction with aging increases risk to insulin resistance and other chronic metabolic diseases. We previously showed functional changes in microRNAs involved in pre-adipocyte differentiation with aging resulting in adipose dysfunction. However, the mechanisms leading to this dysfunction in microRNAs in adipose tissue (adipomiRs) during aging are not well understood. We determined the longitudinal changes in expression of adipomiRs and studied their regulatory mechanisms, such as miRNA biogenesis and editing, in an aging rodent model, with Fischer344 Brown-Norway hybrid rats at ages ranging from 3 to 30 months (male/females, × n > 8). Expression of adipomiRs and their edited forms were determined by small-RNA sequencing. RT-qPCR was used to measure the mRNA expression of biogenesis and editing enzymes. Sanger sequencing was used to validate editing with aging. Differential expression of adipomiRs involved in adipocyte differentiation and insulin signaling was altered with aging. Sex- and age-specific changes in edited adipomiRs were observed. An increase in miRNA biogenesis and editing enzymes (ADARs and their splice variants) were observed with increasing age, more so in female than male rats.
    [Show full text]
  • C9orf72 Frontotemporal Dementia and Amyotrophic Lateral Sclerosis: Investigating Repeat Pathology in Cell Culture Models and Human Post-Mortem Brain
    C9orf72 frontotemporal dementia and amyotrophic lateral sclerosis: investigating repeat pathology in cell culture models and human post-mortem brain. Charlotte Elizabeth Ridler A thesis submitted in partial fulfilment of the requirements for the degree of Doctor of Philosophy from University College London Department of Neurodegenerative Disease Institute of Neurology University College London 2016 1 Declaration I, Charlotte Ridler, confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. 2 Acknowledgements My deepest thanks go to everyone who has helped and supported me during this PhD project, without whom it would not have been possible. Thank you firstly to my supervisor Dr. Adrian Isaacs for all of his guidance, encouragement and enthusiastic discussions during this exciting and unpredictable project. Thank you also to Dr. Sarah Mizielinska, who has been an incredible mentor and role-model during this PhD; I have learnt a huge amount from her endless dedication and generosity. I would also like to thank my secondary supervisor Prof. Elizabeth Fisher for all her wise words of advice. Thank you to all members of the Isaacs and Fisher labs past and present. Special thanks to Frances Norona, Dr. Karen Cleverly, Julian Pietrzyk, Dr. Ione Woollacott and Dr. Anny Devoy who were all great help during the cloning phase of this project; to Dr. Rubika Balendra for all her help and discussions in the nucleoli work; and to Dr. Emma Clayton and Dr. Roberto Simone for their constant support. A number of other people have lent me their valuable expertise for this project, for which I am very grateful.
    [Show full text]
  • View Preprint
    1 Title: Dysregulation of RNA editing may help explain pathogenicity mechanisms of 2 congenital Zika syndrome and Guillain-Barre syndrome 3 4 Helen Piontkivska 1, 2, *, Noel-Marie Plonski 2, Michael M. Miyamoto 3, and Marta L. 5 Wayne 3, 4 6 7 1 Department of Biological Sciences and 2 School of Biomedical Sciences, Kent State University, 8 Kent, OH 44242, USA 9 3 Department of Biology, University of Florida, Gainesville, FL 32611, USA 10 4 Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611, USA 11 12 * Corresponding Author 13 Helen Piontkivska 14 Department of Biological Sciences 15 Kent State University 16 Kent, OH 44242, USA 17 Telephone: (330) 672-3620 18 Fax: (330) 672-3713 19 E-mail: [email protected] 20 1 PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.27401v1 | CC BY 4.0 Open Access | rec: 3 Dec 2018, publ: 3 Dec 2018 21 Abbreviations: 22 ADAR, Adenosine Deaminases Acting on RNA 23 AGS6, Aicardi-Goutieres syndrome 6 24 ALS, amyotrophic lateral sclerosis 25 CDC, Centers for Disease Control and Prevention 26 CDI, inhibitory Ca2+-feedback 27 CMV, cytomegalovirus 28 CZS, Congenital Zika syndrome 29 GBS, Guillain-Barre syndrome 30 IFN, interferon 31 ISGs, interferon-stimulated genes 32 ISREs, interferon-sensitive response elements 33 WNV, West Nile virus 34 Zika virus (ZIKV) 35 2 PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.27401v1 | CC BY 4.0 Open Access | rec: 3 Dec 2018, publ: 3 Dec 2018 36 Abstract: 37 Many Zika virus (ZIKV) pathogenesis-related studies have focused primarily on virus-driven 38 pathology and neurotoxicity, instead of considering the possibility of pathogenesis as an 39 (unintended) consequence of host innate immunity: specifically, as the side-effect of an 40 otherwise well-functioning machine.
    [Show full text]
  • RNA Editing Genes Associated with Extreme Old Age in Humans and with Lifespan in C
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Boston University Institutional Repository (OpenBU) Boston University OpenBU http://open.bu.edu Department of Medicine MED: Medicine Papers 2009-12-14 RNA Editing Genes Associated with Extreme Old Age in Humans and with Lifespan in C. elegans Sebastiani, Paola, Monty Montano, Annibale Puca, Nadia Solovieff, Toshio Kojima, Meng C. Wang, Efthymia Melista, Micah Meltzer, Sylvia E. J. Fischer, Stacy Andersen, Stephen H. Hartley, Amanda Sedgewick, Yasumichi Arai, Aviv Bergman, Nir Barzilai, Dellara F. Terry, Alberto Riva, Chiara Viviani Anselmi, Alberto Malovini, Aya Kitamoto, Motoji Sawabe, Tomio Arai, Yasuyuki Gondo, Martin H. Steinberg, Nobuyoshi Hirose, Gil Atzmon, Gary Ruvkun, Clinton T. Baldwin, Thomas T. Perls. "RNA Editing Genes Associated with Extreme Old Age in Humans and with Lifespan in C. elegans" PLoS ONE 4(12): e8210. (2009) https://hdl.handle.net/2144/2972 Boston University RNA Editing Genes Associated with Extreme Old Age in Humans and with Lifespan in C. elegans Paola Sebastiani1.*, Monty Montano2.*, Annibale Puca3, Nadia Solovieff1, Toshio Kojima4,MengC. Wang5, Efthymia Melista6, Micah Meltzer2, Sylvia E. J. Fischer5, Stacy Andersen7, Stephen H. Hartley1, Amanda Sedgewick8, Yasumichi Arai9, Aviv Bergman10, Nir Barzilai11, Dellara F. Terry7, Alberto Riva12, Chiara Viviani Anselmi3, Alberto Malovini3, Aya Kitamoto4, Motoji Sawabe13, Tomio Arai13, Yasuyuki Gondo14, Martin H. Steinberg6, Nobuyoshi Hirose9, Gil Atzmon11, Gary Ruvkun5, Clinton
    [Show full text]
  • ADAR2 Mislocalization and Widespread RNA Editing Aberrations in C9orf72‑Mediated ALS/FTD
    Acta Neuropathologica https://doi.org/10.1007/s00401-019-01999-w ORIGINAL PAPER ADAR2 mislocalization and widespread RNA editing aberrations in C9orf72‑mediated ALS/FTD Stephen Moore1,2 · Eric Alsop3 · Ileana Lorenzini1 · Alexander Starr1 · Benjamin E. Rabichow1 · Emily Mendez1 · Jennifer L. Levy1 · Camelia Burciu1 · Rebecca Reiman3 · Jeannie Chew4 · Veronique V. Belzil4 · Dennis W. Dickson4 · Janice Robertson5 · Kim A. Staats6 · Justin K. Ichida6 · Leonard Petrucelli4 · Kendall Van Keuren‑Jensen3 · Rita Sattler1 Received: 14 September 2018 / Revised: 28 March 2019 / Accepted: 28 March 2019 © Springer-Verlag GmbH Germany, part of Springer Nature 2019 Abstract The hexanucleotide repeat expansion GGG GCC (G4C2)n in the C9orf72 gene is the most common genetic abnormality asso- ciated with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Recent fndings suggest that dysfunc- tion of nuclear-cytoplasmic trafcking could afect the transport of RNA binding proteins in C9orf72 ALS/FTD. Here, we provide evidence that the RNA editing enzyme adenosine deaminase acting on RNA 2 (ADAR2) is mislocalized in C9orf72 repeat expansion mediated ALS/FTD. ADAR2 is responsible for adenosine (A) to inosine (I) editing of double-stranded RNA, and its function has been shown to be essential for survival. Here we show the mislocalization of ADAR2 in human induced pluripotent stem cell-derived motor neurons (hiPSC-MNs) from C9orf72 patients, in mice expressing (G 4C2)149, and in C9orf72 ALS/FTD patient postmortem tissue. As a consequence of this mislocalization we observe alterations in RNA editing in our model systems and across multiple brain regions. Analysis of editing at 408,580 known RNA editing sites indicates that there are vast RNA A to I editing aberrations in C9orf72-mediated ALS/FTD.
    [Show full text]
  • ADAR2 Induces Reproducible Changes in Sequence and Abundance of Mature Micrornas in the Mouse Brain Cornelia Vesely1, Stefanie Tauber2, Fritz J
    Published online 26 September 2014 Nucleic Acids Research, 2014, Vol. 42, No. 19 12155–12168 doi: 10.1093/nar/gku844 ADAR2 induces reproducible changes in sequence and abundance of mature microRNAs in the mouse brain Cornelia Vesely1, Stefanie Tauber2, Fritz J. Sedlazeck2, Mansoureh Tajaddod1, Arndt von Haeseler2,3 and Michael F. Jantsch1,* 1Department of Chromosome Biology, Max F. Perutz Laboratories, University of Vienna, Dr. Bohr Gasse 9, A-1030 Vienna, Austria, 2Center for Integrative Bioinformatics Vienna, Max F. Perutz Laboratories, University of Vienna, Medical University of Vienna, and University of Veterinary Medicine, Dr. Bohr Gasse 9, A-1030 Vienna, Austria and 3Bioinformatics and Computational Biology, Faculty of Computer Science, University of Vienna, Wahringer¨ Straße 29, A-1090 Vienna, Austria Received July 4, 2014; Revised August 28, 2014; Accepted September 3, 2014 ABSTRACT convert adenosines into inosines in double-stranded RNA structures by hydrolytic deamination. Mammals have three Adenosine deaminases that act on RNA (ADARs) isoforms of ADARs designated as ADAR1 (also known as deaminate adenosines to inosines in double- ADAR), ADAR2 (also known as ADARB1) and ADAR3 stranded RNAs including miRNA precursors. A to I (also known as ADARB2). ADAR3, which is predomi- editing is widespread and required for normal life. By nantly expressed in the brain, is considered to be enzymat- comparing deep sequencing data of brain miRNAs ically inactive as it has no known editing substrates (1). from wild-type and ADAR2 deficient mouse strains, Besides the catalytic deaminase domain, ADARs have two we detect editing sites and altered miRNA processing to three double-stranded RNA binding domains (dsRBDs) at high sensitivity.
    [Show full text]
  • Altered Adenosine-To-Inosine RNA Editing in Human Cancer
    Downloaded from genome.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press Letter Altered adenosine-to-inosine RNA editing in human cancer Nurit Paz,1,2 Erez Y. Levanon,3,12 Ninette Amariglio,1,2 Amy B. Heimberger,4 Zvi Ram,5 Shlomi Constantini,6 Zohar S. Barbash,1,2 Konstantin Adamsky,1 Michal Safran,1,2 Avi Hirschberg,1,2 Meir Krupsky,2,7 Issachar Ben-Dov,2,8 Simona Cazacu,9 Tom Mikkelsen,9 Chaya Brodie,9,10 Eli Eisenberg,11 and Gideon Rechavi1,2,13 1Cancer Research Center, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel; 2Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; 3Compugen Ltd., Tel Aviv 69512, Israel; 4Department of Neurosurgery, Brain Tumor Center, University of Texas M.D. Anderson Cancer Center, Houston 77030, Texas, USA; 5Department of Neurosurgery, Sourasky Medical Center, Tel Aviv 64239, Israel; 6Department of Pediatric Neurosurgery, Dana Children’s Hospital, Sourasky Medical Center, Tel Aviv 64239, Israel; 7Department of Internal Medicine, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel; 8Pulmonary Institute, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel; 9Hermelin Brain Tumor Center, Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan 48202, USA; 10Neuro-Oncology Branch, NCI/NINDS, NIH, Bethesda 20892, Maryland, USA; 11School of Physics and Astronomy, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University 69978 Israel Adenosine-to-inosine (A-to-I) RNA editing was recently shown to be abundant in the human transcriptome, affecting thousands of genes. Employing a bioinformatic approach, we identified significant global hypoediting of Alu repetitive elements in brain, prostate, lung, kidney, and testis tumors.
    [Show full text]
  • Single-Cell RNA Sequencing Reveals Dynamic Changes in A-To-I RNA Editome During Early Human Embryogenesis
    Single-cell RNA sequencing reveals dynamic changes in A-to-I RNA editome during early human embryogenesis Qiu, Si; Li, Wenhui; Xiong, Heng; Liu, Dongbing; Bai, Yali; Wu, Kui; Zhang, Xiuqing; Yang, Huanming; Ma, Kun; Hou, Yong; Li, Bo Published in: BMC Genomics DOI: 10.1186/s12864-016-3115-2 Publication date: 2016 Document version Publisher's PDF, also known as Version of record Document license: CC BY Citation for published version (APA): Qiu, S., Li, W., Xiong, H., Liu, D., Bai, Y., Wu, K., Zhang, X., Yang, H., Ma, K., Hou, Y., & Li, B. (2016). Single- cell RNA sequencing reveals dynamic changes in A-to-I RNA editome during early human embryogenesis. BMC Genomics, 17, [766]. https://doi.org/10.1186/s12864-016-3115-2 Download date: 30. Sep. 2021 Qiu et al. BMC Genomics (2016) 17:766 DOI 10.1186/s12864-016-3115-2 RESEARCH ARTICLE Open Access Single-cell RNA sequencing reveals dynamic changes in A-to-I RNA editome during early human embryogenesis Si Qiu1,2†, Wenhui Li2†, Heng Xiong2, Dongbing Liu2, Yali Bai2,3, Kui Wu2,4, Xiuqing Zhang2, Huanming Yang2,5, Kun Ma2*, Yong Hou2,4* and Bo Li2,6* Abstract Background: A-to-I RNA-editing mediated by ADAR (adenosine deaminase acting on RNA) enzymes that converts adenosine to inosine in RNA sequence can generate mutations and alter gene regulation in metazoans. Previous studies have shown that A-to-I RNA-editing plays vital roles in mouse embryogenesis. However, the RNA-editing activities in early human embryonic development have not been investigated. Results: Here, we characterized genome-wide A-to-I RNA-editing activities during human early embryogenesis by profiling 68 single cells from 29 human embryos spanning from oocyte to morula stages.
    [Show full text]
  • RNA Adenosine Deaminase ADAR2 Modulates T Helper 17 Cell Effector Function
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.22.308221; this version posted September 22, 2020. 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. RNA adenosine deaminase ADAR2 modulates T helper 17 cell effector function Shengyun Ma1,4, Benjamin S. Cho1,4, Yuxin Li1, Nazia Abbasi1, Brian A. Yee1, Ge Sun1, Claire Luo1, John T. Chang2, Bing Zhou1,3, Xiang-dong Fu1, Gene W. Yeo1, Wendy Jia Men Huang1,* 1 Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, U.S.A 2 Department of Medicine, University of California San Diego, La Jolla, CA 92093, U.S.A 3 currently at Institute of Zoology Chinese Academy of Sciences, Beijing 100101, China 4 These authors contributed equally. * Correspondence: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.09.22.308221; this version posted September 22, 2020. 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 Adenosine deaminases acting on RNA (ADARs) catalyze the most common RNA modification in mammals, but it remains to be elucidated how their RNA editing dependent and independent activities contribute to host immunity. Here, we report dynamic changes in ADARs expressions and global adenosine-to-inosin (A-to-I) editome during T helper cell differentiation. In differentiated T helper 17 (Th17) cells, transcription of the ADAR2 encoding locus is potentiated by an intragenic super enhancer and splicing of the ADAR2 encoding transcript is further facilitated by a DEAD-box RNA helicase, DDX5.
    [Show full text]
  • Case-Control Study of ADARB1 and ADARB2 Gene Variants in Migraine
    Gasparini et al. The Journal of Headache and Pain (2015) 16:31 DOI 10.1186/s10194-015-0511-y RESEARCH ARTICLE Open Access Case-control study of ADARB1 and ADARB2 gene variants in migraine Claudia F Gasparini1, Heidi G Sutherland2, Bridget Maher2,3, Astrid J Rodriguez-Acevedo2, Elhame Khlifi2, Larisa M Haupt2 and Lyn R Griffiths2* Abstract Background: Migraine causes crippling attacks of severe head pain along with associated nausea, vomiting, photophobia and/or phonophobia. The aim of this study was to investigate single nucleotide polymorphisms (SNPs) in the adenosine deaminase, RNA-specific, B1 (ADARB1) and adenosine deaminase, RNA specific, B2 (ADARB2) genes in an Australian case–control Caucasian population for association with migraine. Both candidate genes are highly expressed in the central nervous system and fit criteria for migraine neuropathology. SNPs in the ADARB2 gene were previously found to be positively associated with migraine in a pedigree-based genome wide association study using the genetic isolate of Norfolk Island, Australia. The ADARB1 gene was also chosen for investigation due to its important function in editing neurotransmitter receptor transcripts. Methods: Four SNPs in ADARB1 and nine in ADARB2 were selected by inspecting blocks of linkage disequilibrium in Haploview for genotyping using either TaqMan or Sequenom assays. These SNPs were genotyped in two-hundred and ninety one patients who satisfied the International Classification of Headache Disorders-II 2004 diagnostic criteria for migraine, and three-hundred and fourteen controls, and PLINK was used for association testing. Results: Chi-square analysis found no significant association between any of the SNPs tested in the ADARB1 and ADARB2 genes in this study and the occurrence of migraine.
    [Show full text]
  • Chromatin Conformation Links Distal Target Genes to CKD Loci
    BASIC RESEARCH www.jasn.org Chromatin Conformation Links Distal Target Genes to CKD Loci Maarten M. Brandt,1 Claartje A. Meddens,2,3 Laura Louzao-Martinez,4 Noortje A.M. van den Dungen,5,6 Nico R. Lansu,2,3,6 Edward E.S. Nieuwenhuis,2 Dirk J. Duncker,1 Marianne C. Verhaar,4 Jaap A. Joles,4 Michal Mokry,2,3,6 and Caroline Cheng1,4 1Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands; and 2Department of Pediatrics, Wilhelmina Children’s Hospital, 3Regenerative Medicine Center Utrecht, Department of Pediatrics, 4Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, 5Department of Cardiology, Division Heart and Lungs, and 6Epigenomics Facility, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands ABSTRACT Genome-wide association studies (GWASs) have identified many genetic risk factors for CKD. However, linking common variants to genes that are causal for CKD etiology remains challenging. By adapting self-transcribing active regulatory region sequencing, we evaluated the effect of genetic variation on DNA regulatory elements (DREs). Variants in linkage with the CKD-associated single-nucleotide polymorphism rs11959928 were shown to affect DRE function, illustrating that genes regulated by DREs colocalizing with CKD-associated variation can be dysregulated and therefore, considered as CKD candidate genes. To identify target genes of these DREs, we used circular chro- mosome conformation capture (4C) sequencing on glomerular endothelial cells and renal tubular epithelial cells. Our 4C analyses revealed interactions of CKD-associated susceptibility regions with the transcriptional start sites of 304 target genes. Overlap with multiple databases confirmed that many of these target genes are involved in kidney homeostasis.
    [Show full text]