Mechanisms in Ascl1-Mediated Reprogramming of Fibroblasts Into Neurons

Total Page:16

File Type:pdf, Size:1020Kb

Mechanisms in Ascl1-Mediated Reprogramming of Fibroblasts Into Neurons MECHANISMS IN ASCL1-MEDIATED REPROGRAMMING OF FIBROBLASTS INTO NEURONS A DISSERTATION SUBMITTED TO THE DEPARTMENT OF BIOENGINEERING AND THE COMMITTEE ON GRADUATE STUDIES OF STANFORD UNIVERSITY IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY QIAN YI LEE AUGUST 2017 © 2017 by Qian Yi Lee. All Rights Reserved. Re-distributed by Stanford University under license with the author. This work is licensed under a Creative Commons Attribution- Noncommercial 3.0 United States License. http://creativecommons.org/licenses/by-nc/3.0/us/ This dissertation is online at: http://purl.stanford.edu/hf795pb4608 ii I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation for the degree of Doctor of Philosophy. Marius Wernig, Primary Adviser I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation for the degree of Doctor of Philosophy. Russ Altman I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation for the degree of Doctor of Philosophy. Howard Chang I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation for the degree of Doctor of Philosophy. Stephen Quake Approved for the Stanford University Committee on Graduate Studies. Patricia J. Gumport, Vice Provost for Graduate Education This signature page was generated electronically upon submission of this dissertation in electronic format. An original signed hard copy of the signature page is on file in University Archives. iii Abstract Lineage reprogramming of somatic cells, i.e. the conversion of one cell type into another, unrelated cell type, has innovated the fields of stem cell research and translational medicine. The goal of my thesis is to understand the molecular mechanisms of induced lineage reprogramming to improve efficiencies and thereby enable its translation for clinical applications. Previous studies have shown that during reprogramming of fibroblasts into induced pluripotent stem cells (iPSCs) using transcription factors Oct4, Sox2, Klf4 and c-Myc (OSKM), OSK act cooperatively as pioneering factors to first bind fibroblast enchancers to shut down the donor program before moving to activate the pluripotency circuit. It was also shown that iPSC reprogramming involves an initial stochastic phase to activate a subpopulation of cells that eventually enter a late hierarchical phase that activates the endogenous pluripotent circuitry. In contrast, studies in direct reprogramming of fibroblasts into induced neuronal (iN) cells and muscles by Ascl1 and Myod1 respectively have shown that Ascl1 and Myod1 bind immediately to their endogenous binding sites and activate their respective target programs. This leads us to hypothesize that the mechanism of direct reprogramming differs from iPSC reprogramming. Using single cell and bulk RNA sequencing (RNA-seq), Assay of Transposase Accessibly Chromatin with sequencing (ATAC-seq) and Chromatin Immunoprecipitation followed by sequencing (ChIP-seq) to observe Ascl1 binding and changes in gene expression and chromatin accessibility during iN cell reprogramming, we found that in contrast to iPSCs, there is an initial homogenous response to Ascl1 within 2 days of transgene induction. There is a corresponding increase in accessibility of chromatin regions bound by Ascl1 within 12 hours, which leads to the up-regulation of a series of Ascl1 target genes that act in concert to facilitate a major cell fate transition between 2 – 5 days. Finally, a maturation phase between occurs after 5 days whereby the cells begin to activate neuronal and synapse maturation genes to become fully functional neurons. Surprisingly, we also found a small fraction of cells that got redirected to an alternate myogenic program, and showed that it can be attributed to unexpected similarities in Ascl1 and Myod1 DNA-binding affinities. Finally, we found that the myogenic program can by suppressed by pro-neuronal Myt1l to both increase Ascl1- mediated iN cell reprogramming efficiency and also redirect Myod1-expressing cells iv towards a neurogenic fate. These observations underscore the different molecular mechanisms between iN cell and iPSC reprogramming, reveal specific properties of transcription factors able to induce reprogramming, and also show the underlying the importance of endogenous co-factors in regulating cell fate during development. v Acknowledgements I am extremely grateful for the support and encouragement I have received through my PhD career, without which this thesis would not have been possible. I would first like to thank my mentor, Dr. Marius Wernig, for his guidance and support. He always has time to help me brainstorm new ideas, or troubleshoot persisting problems, His passion for science and insightful advice has also pushed me beyond my limits and helped me develop into the scientist I am today. I would like to thank my committee members – Dr. Howard Chang, Dr. Steve Quake and Dr. Russ Altman, as well as my committee chair Dr. Wing Wong, for their invaluable input and career advice. I especially would like thank Dr. Howard Chang for his close mentorship over the years as I collaborated closely with his lab. I would like to thank my other co-authors and collaborators, in particular Dr. Barbara Treutlein, Dr. Orly Wapinski and Dr. Moritz Mall, who have made invaluable contributions to this work, and without their insight and experimental support, this work would not have been possible. I am also thankful to my home department, Bioengineering, especially to Olgalydia and Justin, who had been very helpful and patient with me. Also, my graduate studies would not have been possible without my funding from the Agency of Science, Technology and Research (A*STAR). I would also like to thank everyone in the Wernig lab for their collaboration and support over the years. In particular, I would like to thank the members of our bay, Moritz, Cheen and Justyna for all our discussions, both scientific and not, that helped to push forward new ideas and also make life in lab so much more fun. Thanks also to Nan, Yihan, Sam, Soham and Tommy, who had been a lot of help through the years and been very patient with all my questions, especially when I was still trying to gain my bearings as a new graduate student. And also thank you to everyone (including Bahareh, Bo, Sarah, Lingjun vi and Katie) who was willing to humor me and go out for lunch or dinner or even for a hike to relax away from work. I am also thankful to my Singaporean friends, who formed an amazing and vibrant community here at Stanford. And I am grateful to my family, who have always been there for me and never wavered in their support. And last but not the least, a huge thank you to my boyfriend Winston Koh, who has been steadfast in his support, and has stayed by my side through this long road. vii Table of Contents Abstract ........................................................................................................................ iv Acknowledgements ...................................................................................................... vi Table of Contents ....................................................................................................... viii List of Figures ............................................................................................................... x Introduction .................................................................................................................. 1 REFERENCES ........................................................................................................... 4 CHAPTER 1: Dissecting direct reprogramming from fibroblast to neuron ......... 7 using single-cell RNA-seq ............................................................................................. 7 SUMMARY ................................................................................................................ 8 INTRODUCTION ...................................................................................................... 9 RESULTS ................................................................................................................. 10 EXPERIMENTAL PROCEDURES ......................................................................... 17 ACKNOWLEDGEMENTS ...................................................................................... 25 FIGURE LEGENDS ................................................................................................. 26 FIGURES .................................................................................................................. 30 EXTENDED DATA FIGURE LEGENDS .............................................................. 34 EXTENDED DATA FIGURES ............................................................................... 39 REFERENCES ......................................................................................................... 47 CHAPTER 2: Singular and prescient chromatin switch in the direct reprogramming of fibroblasts to neurons ................................................................ 52 SUMMARY .............................................................................................................. 53 INTRODUCTION .................................................................................................... 54
Recommended publications
  • Prospective Isolation of NKX2-1–Expressing Human Lung Progenitors Derived from Pluripotent Stem Cells
    The Journal of Clinical Investigation RESEARCH ARTICLE Prospective isolation of NKX2-1–expressing human lung progenitors derived from pluripotent stem cells Finn Hawkins,1,2 Philipp Kramer,3 Anjali Jacob,1,2 Ian Driver,4 Dylan C. Thomas,1 Katherine B. McCauley,1,2 Nicholas Skvir,1 Ana M. Crane,3 Anita A. Kurmann,1,5 Anthony N. Hollenberg,5 Sinead Nguyen,1 Brandon G. Wong,6 Ahmad S. Khalil,6,7 Sarah X.L. Huang,3,8 Susan Guttentag,9 Jason R. Rock,4 John M. Shannon,10 Brian R. Davis,3 and Darrell N. Kotton1,2 2 1Center for Regenerative Medicine, and The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA. 3Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA. 4Department of Anatomy, UCSF, San Francisco, California, USA. 5Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA. 6Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA. 7Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA. 8Columbia Center for Translational Immunology & Columbia Center for Human Development, Columbia University Medical Center, New York, New York, USA. 9Department of Pediatrics, Monroe Carell Jr. Children’s Hospital, Vanderbilt University, Nashville, Tennessee, USA. 10Division of Pulmonary Biology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA. It has been postulated that during human fetal development, all cells of the lung epithelium derive from embryonic, endodermal, NK2 homeobox 1–expressing (NKX2-1+) precursor cells.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • E Proteins Sharpen Neurogenesis by Modulating Proneural Bhlh
    RESEARCH ARTICLE E proteins sharpen neurogenesis by modulating proneural bHLH transcription factors’ activity in an E-box-dependent manner Gwenvael Le Dre´ au1†*, Rene´ Escalona1†‡, Raquel Fueyo2, Antonio Herrera1, Juan D Martı´nez1, Susana Usieto1, Anghara Menendez3, Sebastian Pons3, Marian A Martinez-Balbas2, Elisa Marti1 1Department of Developmental Biology, Instituto de Biologı´a Molecular de Barcelona, Barcelona, Spain; 2Department of Molecular Genomics, Instituto de Biologı´a Molecular de Barcelona, Barcelona, Spain; 3Department of Cell Biology, Instituto de Biologı´a Molecular de Barcelona, Barcelona, Spain Abstract Class II HLH proteins heterodimerize with class I HLH/E proteins to regulate transcription. Here, we show that E proteins sharpen neurogenesis by adjusting the neurogenic strength of the distinct proneural proteins. We find that inhibiting BMP signaling or its target ID2 in *For correspondence: the chick embryo spinal cord, impairs the neuronal production from progenitors expressing [email protected] ATOH1/ASCL1, but less severely that from progenitors expressing NEUROG1/2/PTF1a. We show this context-dependent response to result from the differential modulation of proneural proteins’ †These authors contributed activity by E proteins. E proteins synergize with proneural proteins when acting on CAGSTG motifs, equally to this work thereby facilitating the activity of ASCL1/ATOH1 which preferentially bind to such motifs. Present address: Conversely, E proteins restrict the neurogenic strength of NEUROG1/2 by directly inhibiting their ‡ Departamento de Embriologı´a, preferential binding to CADATG motifs. Since we find this mechanism to be conserved in Facultad de Medicina, corticogenesis, we propose this differential co-operation of E proteins with proneural proteins as a Universidad Nacional Auto´noma novel though general feature of their mechanism of action.
    [Show full text]
  • Transcription Factor ATF5 Is Required for Terminal Differentiation and Survival of Olfactory Sensory Neurons
    Transcription factor ATF5 is required for terminal differentiation and survival of olfactory sensory neurons Shu-Zong Wanga,b, Jianhong Oub, Lihua J. Zhub,c, and Michael R. Greena,b,1 aHoward Hughes Medical Institute, bPrograms in Gene Function and Expression and Molecular Medicine, and cProgram in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605 Edited by Solomon H. Snyder, The Johns Hopkins University School of Medicine, Baltimore, MD, and approved September 27, 2012 (received for review June 21, 2012) Activating transcription factor 5 (ATF5) is a member of the ATF/cAMP not been determined. Here we address this issue by deriving and −/− response element-binding family of transcription factors, which characterizing Atf5 mice. compose a large group of basic region leucine zipper proteins whose members mediate diverse transcriptional regulatory functions. Results −/− ATF5 has a well-established prosurvival activity and has been found Derivation of Atf5 Mice. To investigate the physiological role of −/− to be overexpressed in several human cancers, in particular glioblas- ATF5, we derived Atf5 mice. The gene-targeting strategy in- toma. However, the role(s) of ATF5 in development and normal volved replacing almost the entire coding region of Atf5 with a physiology are unknown. Here we address this issue by deriving LacZ-Neo cassette, such that the LacZ gene is under the control and characterizing homozygous Atf5 knockout mice. We find that of the endogenous Atf5 promoter (Fig. S1A). The genomic struc- +/+ +/− −/− Atf5−/− pups die neonatally, which, as explained below, is consis- ture of the Atf5 locus in Atf5 , Atf5 ,andAtf5 mice was fi tent with an olfactory defect resulting in a competitive suckling con rmed by Southern blot (Fig.
    [Show full text]
  • Molecular Regulation in Dopaminergic Neuron Development
    International Journal of Molecular Sciences Review Molecular Regulation in Dopaminergic Neuron Development. Cues to Unveil Molecular Pathogenesis and Pharmacological Targets of Neurodegeneration Floriana Volpicelli 1 , Carla Perrone-Capano 1,2 , Gian Carlo Bellenchi 2,3, Luca Colucci-D’Amato 4,* and Umberto di Porzio 2 1 Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; fl[email protected] (F.V.); [email protected] (C.P.C.) 2 Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, CNR, 80131 Rome, Italy; [email protected] (G.C.B.); [email protected] (U.d.P.) 3 Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy 4 Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy * Correspondence: [email protected]; Tel.: +39-0823-274577 Received: 28 April 2020; Accepted: 1 June 2020; Published: 3 June 2020 Abstract: The relatively few dopaminergic neurons in the mammalian brain are mostly located in the midbrain and regulate many important neural functions, including motor integration, cognition, emotive behaviors and reward. Therefore, alteration of their function or degeneration leads to severe neurological and neuropsychiatric diseases. Unraveling the mechanisms of midbrain dopaminergic (mDA) phenotype induction and maturation and elucidating the role of the gene network involved in the development and maintenance of these neurons is of pivotal importance to rescue or substitute these cells in order to restore dopaminergic functions. Recently, in addition to morphogens and transcription factors, microRNAs have been identified as critical players to confer mDA identity. The elucidation of the gene network involved in mDA neuron development and function will be crucial to identify early changes of mDA neurons that occur in pre-symptomatic pathological conditions, such as Parkinson’s disease.
    [Show full text]
  • Notch Controls Multiple Pancreatic Cell Fate Regulators Through Direct Hes1-Mediated Repression
    bioRxiv preprint doi: https://doi.org/10.1101/336305; this version posted June 1, 2018. 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. Notch Controls Multiple Pancreatic Cell Fate Regulators Through Direct Hes1-mediated Repression by Kristian H. de Lichtenberg1, Philip A. Seymour1, Mette C. Jørgensen1, Yung-Hae Kim1, Anne Grapin-Botton1, Mark A. Magnuson2, Nikolina Nakic3,4, Jorge Ferrer3, Palle Serup1* 1 Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, Denmark. 2 Center for Stem Cell Biology, Vanderbilt University, Tennessee, USA. 3 Section on Epigenetics and Disease, Imperial College London, UK. 4 Current Address: GSK, Stevenage, UK. *Corresponding Author: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/336305; this version posted June 1, 2018. 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. Abstract Notch signaling and its effector Hes1 regulate multiple cell fate choices in the developing pancreas, but few direct target genes are known. Here we use transcriptome analyses combined with chromatin immunoprecipitation with next-generation sequencing (ChIP-seq) to identify direct target genes of Hes1. ChIP-seq analysis of endogenous Hes1 in 266-6 cells, a model of multipotent pancreatic progenitor cells, revealed high-confidence peaks associated with 354 genes. Among these were genes important for tip/trunk segregation such as Ptf1a and Nkx6-1, genes involved in endocrine differentiation such as Insm1 and Dll4, and genes encoding non-pancreatic basic-Helic-Loop-Helix (bHLH) factors such as Neurog2 and Ascl1.
    [Show full text]
  • Blocking Notch-Signaling Increases Neurogenesis in the Striatum After Stroke
    cells Communication Blocking Notch-Signaling Increases Neurogenesis in the Striatum after Stroke 1 1, 2 2 Giuseppe Santopolo , Jens P. Magnusson y, Olle Lindvall , Zaal Kokaia and Jonas Frisén 1,* 1 Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden; [email protected] (G.S.); [email protected] (J.P.M.) 2 Lund Stem Cell Center, University Hospital, SE-221 84 Lund, Sweden; [email protected] (O.L.); [email protected] (Z.K.) * Correspondence: [email protected] Current affiliation: Department of Bioengineering, Stanford University, Stanford, CA 94305, USA. y Received: 13 June 2020; Accepted: 16 July 2020; Published: 20 July 2020 Abstract: Stroke triggers neurogenesis in the striatum in mice, with new neurons deriving in part from the nearby subventricular zone and in part from parenchymal astrocytes. The initiation of neurogenesis by astrocytes within the striatum is triggered by reduced Notch-signaling, and blocking this signaling pathway by deletion of the gene encoding the obligate Notch coactivator Rbpj is sufficient to activate neurogenesis by striatal astrocytes in the absence of an injury. Here we report that blocking Notch-signaling in stroke increases the neurogenic response to stroke 3.5-fold in mice. Deletion of Rbpj results in the recruitment of a larger number of parenchymal astrocytes to neurogenesis and over larger areas of the striatum. These data suggest inhibition of Notch-signaling as a potential translational strategy to promote neuronal regeneration after stroke. Keywords: neurogenesis; astrocyte; stem cell; stroke; striatum 1. Introduction Every year, 13.7 million people worldwide suffer a stroke, of whom 5.5 million die and as many suffer permanent loss of function with debilitating outcomes [1].
    [Show full text]
  • Identification of Porcine RUNX1 As an LPS-Dependent Gene Expression Regulator
    bioRxiv preprint doi: https://doi.org/10.1101/713206; this version posted July 24, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1 Identification of porcine RUNX1 as an LPS-dependent gene expression regulator 2 in PBMCs by Super deepSAGE sequencing of multiple tissues 3 Tinghua Huang1, Min Yang1, Kaihui Dong1, Mingjiang Xu1, Jinhui Liu1, Zhi Chen1, 4 Shijia Zhu1, Wang Chen1, Jun Yin1, Kai Jin1, Yu Deng1, Zhou Guan1, Xiali Huang1, 5 Jun Yang1, Rongxun Han1, Min Yao1 6 1. College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China 7 Corresponding author's contact information: College of Animal Science, Yangtze 8 University, Jingzhou, Hubei 434025, China, Email: [email protected], Tel: 9 8613387202183 10 Running title: RUNX1 is a LPS-dependent regulator in PBMCs 11 Keywords: RUNX1, Super deepSAGE, PBMC, LPS 12 13 14 15 16 17 18 19 20 21 1 / 31 bioRxiv preprint doi: https://doi.org/10.1101/713206; this version posted July 24, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 22 Abstract 23 Genome-wide identification of gene expression regulators may facilitate our 24 understanding of the transcriptome constructed by gene expression profiling 25 experiment.
    [Show full text]
  • 1-S2.0-S0959438816302392-Main
    Beyond proneural: emerging functions and regulations of proneural proteins François Guillemot, Bassem A. Hassan To cite this version: François Guillemot, Bassem A. Hassan. Beyond proneural: emerging functions and regula- tions of proneural proteins. Current Opinion in Neurobiology, Elsevier, 2017, 42, pp.93 - 101. 10.1016/j.conb.2016.11.011. hal-01434997 HAL Id: hal-01434997 https://hal.sorbonne-universite.fr/hal-01434997 Submitted on 13 Jan 2017 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Distributed under a Creative Commons Attribution - NonCommercial - NoDerivatives| 4.0 International License Available online at www.sciencedirect.com ScienceDirect Beyond proneural: emerging functions and regulations of proneural proteins 1 2 Franc¸ois Guillemot and Bassem A Hassan Proneural proteins, which include Ascl1, Atoh1 and in multipotent cells and a role in fate specification distin- Neurogenins e´ pinie` re in vertebrates and Achaete-Scute guish proneural proteins from neuronal differentiation proteins and Atonal in Drosophila, are expressed in the factors such as members of the mammalian Neurod developing nervous system throughout the animal kingdom family, which are first expressed in cells already commit- and have an essential and well-characterised role in specifying ted to a neuronal fate and promote their differentiation the neural identity of progenitors.
    [Show full text]
  • Primary Cilia Regulate Hippocampal Neurogenesis by Mediating Sonic Hedgehog Signaling
    Primary cilia regulate hippocampal neurogenesis by mediating sonic hedgehog signaling Joshua J. Breunig*†, Matthew R. Sarkisian*†, Jon I. Arellano*, Yury M. Morozov*, Albert E. Ayoub*, Sonal Sojitra‡, Baolin Wang§¶, Richard A. Flavellʈ**, Pasko Rakic*††, and Terrence Town‡ʈ *Department of Neurobiology and Kavli Institute of Neuroscience and §Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10021; ¶Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10021; ʈDepartment of Immunobiology and **Howard Hughes Medical Institute, Yale University School of Medicine, 300 Cedar Street, TAC S-569, New Haven, CT 06519-8011; and ‡Departments of Biomedical Science, Neurosurgery, and Medicine, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048 Contributed by Pasko Rakic, May 10, 2008 (sent for review March 20, 2008) Primary cilia are present on mammalian neurons and glia, but their tivity, we noted that the typical lamination and morphology of function is largely unknown. We generated conditional homozy- glial cells was severely disrupted (Fig. 1 E–H). Glial cell bodies, gous mutant mice for a gene we termed Stumpy. Mutants lack cilia as shown by combined Sox2 and Gfap immunoreactivity, were and have conspicuous abnormalities in postnatally developing located more randomly in NestinCre; Stumpyfl/fl mice (hereto- brain regions, including a hypoplasic hippocampus characterized fore referred to as ⌬Stumpy mice), and there was a notable lack by a primary deficiency in neural stem cells known as astrocyte-like of DG radial glia (Fig. 1 A, B, and E–H).
    [Show full text]
  • Highly Dynamic Chromatin Interactions Drive Neurogenesis Through Gene
    bioRxiv preprint doi: https://doi.org/10.1101/303842; this version posted April 18, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Highly dynamic chromatin interactions drive neurogenesis through gene regulatory networks Valeriya Malysheva1,2,4, Marco Antonio Mendoza-Parra1, Matthias Blum1,3 and Hinrich Gronemeyer1,4 1 Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Equipe Labellisée Ligue Contre le Cancer, Centre National de la Recherche Scientifique UMR 7104, Institut National de la Santé et de la Recherche Médicale U964, University of Strasbourg, Illkirch, France. 2 Present address: The Babraham Institute, Babraham Research Campus, Cambridge, UK 3 Present address: European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, UK 4 Corresponding authors: Valeriya Malysheva Email: [email protected] Phone: +(44) 1223 49 6505 Hinrich Gronemeyer Email: [email protected] Phone: +(33) 3 88 65 34 73 bioRxiv preprint doi: https://doi.org/10.1101/303842; this version posted April 18, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Cell fate transitions are fundamental processes in the ontogeny of multicellular organisms1 and aberrations can generate pathologies2. Cell fate acquisition is a highly complex phenomenon which involves a plethora of intrinsic and extrinsic instructive signals that direct the lineage progression of pluripotent cells.
    [Show full text]
  • The Hormone-Bound Vitamin D Receptor Regulates Turnover of Target
    The hormone-bound vitamin D receptor regulates turnover of target proteins of the SCFFBW7 E3 ligase By Reyhaneh Salehi Tabar Department of Experimental Medicine McGill University Montreal, QC, Canada April 2016 A thesis submitted to McGill University in partial fulfillment of the requirements of the degree of Doctor of Philosophy © Reyhaneh Salehi Tabar 1 Table of Contents Abbreviations ................................................................................................................................................ 7 Abstract ....................................................................................................................................................... 10 Rèsumè ....................................................................................................................................................... 13 Acknowledgements ..................................................................................................................................... 16 Preface ........................................................................................................................................................ 17 Contribution of authors .............................................................................................................................. 18 Chapter 1-Literature review........................................................................................................................ 20 1.1. General introduction and overview of thesis ............................................................................
    [Show full text]