Regulation of A2 Chimaerin and Associated Phosphorylation Pathways in Neuronal Signalling and Morphogenesis

Total Page:16

File Type:pdf, Size:1020Kb

Regulation of A2 Chimaerin and Associated Phosphorylation Pathways in Neuronal Signalling and Morphogenesis Regulation of a2 Chimaerin and Associated Phosphorylation Pathways in Neuronal Signalling and Morphogenesis By Tom Jacobs Submitted to University College London for the degree of Doctor of Philosophy, Ph.D. 2004 Department of Molecular Neuroscience Institute of Neurology University College London 1 UMI Number: U602542 All rights reserved INFORMATION TO ALL USERS The quality of this reproduction is dependent upon the quality of the copy submitted. In the unlikely event that the author did not send a complete manuscript and there are missing pages, these will be noted. Also, if material had to be removed, a note will indicate the deletion. Dissertation Publishing UMI U602542 Published by ProQuest LLC 2014. Copyright in the Dissertation held by the Author. Microform Edition © ProQuest LLC. All rights reserved. This work is protected against unauthorized copying under Title 17, United States Code. ProQuest LLC 789 East Eisenhower Parkway P.O. Box 1346 Ann Arbor, Ml 48106-1346 ^Uo mp darling wife, ^fulie, fo r pour constant encouragement and support. ^Uo mp parents, fo r alwaps being there and for helping to get me here Acknowledgements I would like to thank Prof. Louis Lim not only for the opportunity to study for a PhD in his London lab, but also for the fantastic experience of working in Singapore and not to mention, allowing me to meet my wife, Julie. To my supervisor Dr. Chrisine Hall, thank you for all your guidance and encouragement during the course of this work. Thanks to Dr. Clinton Monfiies for the use of numerous molecular biology reagents, Dr Kate Marler and Dr Sheila Govind for initial demonstrations and training and Mathew Brown for insightful discussions. Thank you also to lab members Bhav and Sally for all your help and assistance but especially for cheering up the working environment. Thank you to all members of the Singapore lab for making me feel so welcome during my stay. Special thanks to my wife Dr Julie Edgeworth for all your emotional and professional support, encouragement and much needed motivation. Thanks to my parents, who have always supported me and put me in the position to do a PhD in the first place. 3 Abstract Abstract The generation of neuronal dendrites and axons is a highly dynamic process. These extensive structural changes are regulated by molecules such as GTPases, as well as kinases/phosphatases, which provide cyclical control mechanisms. Rho family GTPases are regulated, in part, by GAPs such as neurone-enriched chimaerin, and act through downstream kinase effectors, including cyclin dependant kinase 5 (Cdk5). a 2 chimaerin is a Rac GAP and phorbol ester receptor, which also displays Cdc42/Rac effector functions. From this study it appears a2 chimaerin exists in an auto- inhibited state in the cytosol and that DAG/phorbol ester association, or selected mutations translocates a2 chimaerin to the membrane, leading to GAP activation. a2 chimaerin GAP activity was implicated in PMA induced neurite collapse of N1E-115 neuroblastoma cells. PMA also promoted the association of a2 chimaerin with target proteins Collapsin Response Mediator Protein-2 (CRMP-2) and p35/Cdk5, possibly as a result of a conformational change. The Rac effector, Cdk5 is a proline directed, Ser/Thr kinase, whose activity contributes to neurite outgrowth, growth cone collapse and neuronal migration. Deregulation of Cdk5 in neurodegenerative diseases is implicated in pathological changes. a2-chimaerin GAP domain was shown to interact in vivo both with p35, the neuronal activator of Cdk5 and with Cdk5 itself. CRMP-2 interacted in vivo with a2 chimaerin SH2 domain, for which tyrosine phosphorylation was not essential. The functional correlates of these protein interactions of a2 chimaerin were investigated in vivo. CRMP-2, which is hyperphosphorylated in Alzheimer’s disease was identified as a novel Cdk5 substrate in vitro with CRMP-2 Ser522 identified as the target site. Phosphorylation of CRMP-2 Ser522, potentially primes for GSK-3P phosphorylation of CRMP-2 Ser518 and Thr 514 which alters CRMP-2 conformation. a2 chimaerin was not a Cdk5 substrate but may be tyrosine phosphorylated, which could potentially regulate its activity or associations in neuronal cells. 4 Table of Contents TABLE OF CONTENTS Title Page 1 Dedication 2 Acknowledgements 3 Abstract 4 Table of Contents 5-10 List of Figures 11-14 Abbreviations 15-18 Chapter 1 Introduction 19-54 1A The Cytoskeleton 20-25 1.A.1 Microtubules 21-22 1.A3 Actin Filaments 22-25 1 A.2.1 Actin binding proteins 22-23 l.A.2.2 Actin filament nucleation 23-24 IA.2.3 Actin filament depolymerisation 24 l.A.2.4 Actin filament organisation 25 1.B Rho GTPases 26-34 1.B.1 Guanine nucleotide exchange factors (GEFs) 27-28 I.B.2 GTPase activating proteins (GAPs) 28-33 IB.2.1 Rho GAP regulation ' 29-54 1 B.2.1.1 Lipid association 29-30 l.B.2.1.2 Protein interactions 30-31 1 .B.2.1.3 Phosphorylation 31 -32 l.B.2.2 Rho GAP effector functions 32-33 1.B3 Guanine nucleotide dissociation inhibitors (GDI) 33 1.B.4 Alternative mechanisms of Rho GTPase regulation 33-34 l.C The Rho GTPases and the cytoskeleton 34-37 l.C .l Actin dynamics 34-36 1.C2 Microtubule dynamics 36-37 5 Table of Contents 1.D Central Nervous System (CNS) Development 37-54 1.D.1 Neuronal Migration 38-40 l.D.1.1 Cyclin dependent kinase 5 (Cdk5) 38-40 1JX2 Neurite outgrowth 40-47 l.D.2.1 Rho GAPs 42-43 1.D 22 Collapsin response mediator protein 2 (CRMP-2) 43-44 l.D.2.3 Cyclin dependent kinase 5 (Cdk5) 45-47 1J>3 Axonal Guidance 47- 52 l.D.3.1 Semaphorins 49-51 l.D.3.2 Regulation of guidance cues 51-52 1.D.4 Neurodegenerative Diseases 52-54 1.E Aims 54 Chanter 2 Materials & Methods 55-78 2.A Materials 56-58 2A.1 Laboratory Reagents 56 2.A.2 DNA Manipulation Reagents 56 2.A3 Tissue Culture Reagents 56 2A.4 Protein Work Reagents 57 2 .A 3 Antibodies 57 2.A.6 cDNA Constructs 58 2.A.7 Oligomer Synthesis ans cDNA sequenecing 58 2.B Methods 58-78 2.B.1 DNA Manipulation 58-66 2.B.1.1 Competent bacterial transformation 58 2.B.1.2 Plasmid DNA mini preparations 59-60 2.B.1.3 Plasmid DNA midi preparations 60-61 2.B.1.4 Phenol/Chloroform extraction 61 2.B.1.5 Ethanol precipitation 62 2.B.1.6 Quantitation of DNA 62 2.B.1.7 Restriction digestion of DNA 62 2.B.1.8 Agarose gel electrophoresis 63 6 Table of Contents 2.B. 1.9 Visualisation of DNA with ethidium bromide 63 2.B.1.10 Site directed mutagenesis 63-65 2.B. 1.11 Identification of positive mutant clones 66 23.2 Cell Culture 67 23.2.1 Cell stocks 67 233 Morphological Analysis 68-70 23.3.1 Coverslip preparation 68 2.B.3.2 Transfection on coverslips 68 2.B.3.3 Immunostaining 68-69 2.B.3.4 Fixed cell imaging 69 2.B.3.5 Live cell imaging 70 2.B.4 Protein W ork 70-75 23.4.1 Recombinant protein production 70-71 2.B.4.1.1 GST Fused 71 23.4.13 Non Tagged Protein 71 23.4.2 Mammalian cell90mm plate transfection 71 2.B.4.3 Cell treatment 72 2.B.4.3.1 Pervanadate 72 2.B.4.3.2 Phorbol 12-myristate 13-acetate (PMA) 72 2.B.4.4 Cell lysis 72-73 23.4.5 Immunoprecipitation/GST-pulldown 73 2.B.4.6 Rac-GTP assay 74 2.B.4.7 Cell fiactionation 74-75 2.B.4.8 In vitro kinase assays 75 2.B5 Protein Analysis 75-78 2.B.5.1 Protein quantification 75 23.5.2 SDS-polyacrylamide gel electrophoresis 76-77 2.B.5.3 Commassie blue staining 77 2.B.5.4 Western transfer (semi dry blotting) 77 2.B.5.5 Immunoblotting 78 7 Table of Contents Chapter 3 Results I 79-101 (x2 Chimaerin: GAP Regulation and Neuronal Morphology 80 3.A a2 Chimaerin Location 81-85 3.A.1 Mutations in the SH2 domain and GAP domain of a2 Chimaerin alter subcellular distribution 81-83 3A.2 PMA translocates a2 chimaerin 83-85 3.B a2 Chimaerin GAP Activity and Neuroblastoma Cell Morphology 86-96 3.B.1 PMA causes retraction of a2.10 neuroblastoma cells accompanied by a decrease in Rac-GTP 86-88 3 B 2 PMA causes retraction of N1E-115 neuroblastoma cells which can be inhibited with the expression of GAP inactive a 2 chimaerin 88-92 3.B.3 a2 Chimaerin GAP activity inhibits neuritogenesis of N1E-115 neuroblastoma cells when membrane targeted 92-94 3.B.4 GAP inactive a2 Chimaerin expression induces neurite outgrowth inNlE-115 neuroblastoma cells 94-96 3.C PMA Activates a2 Chimaerin GAP activity 96-100 3.C.1 PMA alters a2 chimaerin distribution in Cos-7 Cells 96-98 3.C.2 a2 Chimaerin inhibits PMA induced lamellipodia in Cos-7 Cells 98-100 3.D Summary of Results I 100-101 Chapter 4 Results II 102-126 a2 Chimaerin and P35/Cdk5 103 4A.1 p35 interacts with a2 Chimaerin C-terminus 104 4.A.2 a2 Chimaerin is not an in vitro p25/Cdk5 substrate 105-106 4.A.3 a2 Chimaerin interacts separately with p35 and Cdk5, but not with the active p35/Cdk5 complex 106-109 4A.4 a2 Chimaerin does not regulate p35/Cdk5 in vitro 109-110 4A.5 PMA promotes o2 Chimaerin binding with p35 111-112 8 Table of Contents 4A.6 <x2 Chimaerin C-terminal gap domain is sufficient for Cdk5 association 112-114 4A.7 PMA increases binding of a2 Chimaerin with p35/Cdk5 114-118 4.A.8 p35 or CRMP-2 co-expression does not regulate a2 chimaerin induced morphology 118-121 4A.9 p35 expression in neuroblastoma cells dissociates a 2 chimaerin from the membrane fraction 122-123 4A.10 Cdk5 Kinase activity is not required for neuritogenesis of N1E-115 neuroblastoma cells 123-125 4.B Summary of Results II 125-126 Chapter 5 Results III 127-160 A2 Chimaerin SH2 Domain and Associated Phosphorylation Pathways 128 5.A Phosphotyrosine binding is not required for GAP inactive a2 Chimaerin induction of neurites 129-130 5.B a2 Chimaerin and CRMP-2 130-171 5.B.1 PMA increases CRMP-2 association with a2 chimaerin in vivo 130-132 5.B.2 a2
Recommended publications
  • Adaptive Stress Signaling in Targeted Cancer Therapy Resistance
    Oncogene (2015) 34, 5599–5606 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc REVIEW Adaptive stress signaling in targeted cancer therapy resistance E Pazarentzos1,2 and TG Bivona1,2 The identification of specific genetic alterations that drive the initiation and progression of cancer and the development of targeted drugs that act against these driver alterations has revolutionized the treatment of many human cancers. Although substantial progress has been achieved with the use of such targeted cancer therapies, resistance remains a major challenge that limits the overall clinical impact. Hence, despite progress, new strategies are needed to enhance response and eliminate resistance to targeted cancer therapies in order to achieve durable or curative responses in patients. To date, efforts to characterize mechanisms of resistance have primarily focused on molecular events that mediate primary or secondary resistance in patients. Less is known about the initial molecular response and adaptation that may occur in tumor cells early upon exposure to a targeted agent. Although understudied, emerging evidence indicates that the early adaptive changes by which tumor cells respond to the stress of a targeted therapy may be crucial for tumo r cell survival during treatment and the development of resistance. Here we review recent data illuminating the molecular architecture underlying adaptive stress signaling in tumor cells. We highlight how leveraging this knowledge could catalyze novel strategies to minimize
    [Show full text]
  • The Atypical Guanine-Nucleotide Exchange Factor, Dock7, Negatively Regulates Schwann Cell Differentiation and Myelination
    The Journal of Neuroscience, August 31, 2011 • 31(35):12579–12592 • 12579 Cellular/Molecular The Atypical Guanine-Nucleotide Exchange Factor, Dock7, Negatively Regulates Schwann Cell Differentiation and Myelination Junji Yamauchi,1,3,5 Yuki Miyamoto,1 Hajime Hamasaki,1,3 Atsushi Sanbe,1 Shinji Kusakawa,1 Akane Nakamura,2 Hideki Tsumura,2 Masahiro Maeda,4 Noriko Nemoto,6 Katsumasa Kawahara,5 Tomohiro Torii,1 and Akito Tanoue1 1Department of Pharmacology and 2Laboratory Animal Resource Facility, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan, 3Department of Biological Sciences, Tokyo Institute of Technology, Midori, Yokohama 226-8501, Japan, 4IBL, Ltd., Fujioka, Gumma 375-0005, Japan, and 5Department of Physiology and 6Bioimaging Research Center, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan In development of the peripheral nervous system, Schwann cells proliferate, migrate, and ultimately differentiate to form myelin sheath. In all of the myelination stages, Schwann cells continuously undergo morphological changes; however, little is known about their underlying molecular mechanisms. We previously cloned the dock7 gene encoding the atypical Rho family guanine-nucleotide exchange factor (GEF) and reported the positive role of Dock7, the target Rho GTPases Rac/Cdc42, and the downstream c-Jun N-terminal kinase in Schwann cell migration (Yamauchi et al., 2008). We investigated the role of Dock7 in Schwann cell differentiation and myelination. Knockdown of Dock7 by the specific small interfering (si)RNA in primary Schwann cells promotes dibutyryl cAMP-induced morpholog- ical differentiation, indicating the negative role of Dock7 in Schwann cell differentiation. It also results in a shorter duration of activation of Rac/Cdc42 and JNK, which is the negative regulator of myelination, and the earlier activation of Rho and Rho-kinase, which is the positive regulator of myelination.
    [Show full text]
  • A Novel Method of Neural Differentiation of PC12 Cells by Using Opti-MEM As a Basic Induction Medium
    INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 41: 195-201, 2018 A novel method of neural differentiation of PC12 cells by using Opti-MEM as a basic induction medium RENDONG HU1*, QIAOYU CAO2*, ZHONGQING SUN3, JINYING CHEN4, QING ZHENG2 and FEI XIAO1 1Department of Pharmacology, School of Medicine, Jinan University; 2College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632; 3Department of Anesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, SAR; 4Department of Ophthalmology, The First Clinical Medical College of Jinan University, Guangzhou, Guangdong 510632, P.R. China Received April 5, 2017; Accepted October 11, 2017 DOI: 10.3892/ijmm.2017.3195 Abstract. The PC12 cell line is a classical neuronal cell model Introduction due to its ability to acquire the sympathetic neurons features when deal with nerve growth factor (NGF). In the present study, The PC12 cell line is traceable to a pheochromocytoma from the authors used a variety of different methods to induce PC12 the rat adrenal medulla (1-4). When exposed to nerve growth cells, such as Opti-MEM medium containing different concen- factor (NGF), PC12 cells present an observable change in trations of fetal bovine serum (FBS) and horse serum compared sympathetic neuron phenotype and properties. Neural differ- with RPMI-1640 medium, and then observed the neurite length, entiation of PC12 has been widely used as a neuron cell model differentiation, adhesion, cell proliferation and action poten- in neuroscience, such as in the nerve injury-induced neuro- tial, as well as the protein levels of axonal growth-associated pathic pain model (5) and nitric oxide-induced neurotoxicity protein 43 (GAP-43) and synaptic protein synapsin-1, among model (6).
    [Show full text]
  • Agrin Differentially Regulates the Rates of Axonal and Dendritic Elongation in Cultured Hippocampal Neurons
    The Journal of Neuroscience, September 1, 2001, 21(17):6802–6809 Agrin Differentially Regulates the Rates of Axonal and Dendritic Elongation in Cultured Hippocampal Neurons Kristina B. Mantych and Adriana Ferreira Institute for Neuroscience and Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611 In the present study, we examined the role of agrin in axonal elongation and branching were paralleled by changes in the and dendritic elongation in central neurons. Dissociated hip- composition of the cytoskeleton. In the presence of agrin, there pocampal neurons were grown in the presence of either recom- was an upregulation of the expression of microtubule- binant agrin or antisense oligonucleotides designed to block associated proteins MAP1B, MAP2, and tau. In contrast, a agrin expression. Our results indicate that agrin differentially downregulation of the expression of these MAPs was detected regulates axonal and dendritic growth. Recombinant agrin de- in agrin-depleted cells. Taken collectively, these results suggest creased the rate of elongation of main axons but induced the an important role for agrin as a trigger of the transcription of formation of axonal branches. On the other hand, agrin induced neuro-specific genes involved in neurite elongation and branch- both dendritic elongation and dendritic branching. Conversely, ing in central neurons. cultured hippocampal neurons depleted of agrin extended longer, nonbranched axons and shorter dendrites when com- Key words: agrin; neurite outgrowth; microtubule-associated pared with controls. These changes in the rates of neurite proteins; CREB; antisense oligonucleotides; axons and dendrites Agrin, an extracellular matrix protein, is synthesized by motor Conversely, neurons depleted of agrin elongated longer axons neurons and transported to their terminals where it is released when compared with control ones (Ferreira 1999; Serpinskaya et (Magill-Solc and McMahan, 1988; Martinou et al., 1991; Ruegg et al., 1999).
    [Show full text]
  • Mutation Analysis of Son of Sevenless in Juvenile Myelomonocytic Leukemia
    Letters to the Editor 1108 3 Licht JD. Reconstructing a disease: what essential features of the the development of acute promyelocytic leukemia in transgenic retinoic acid receptor fusion oncoproteins generate acute promye- mice. Proc Natl Acad Sci USA 2000; 97: 13306–13311. locytic leukemia? Cancer Cell 2006; 9: 73–74. 10 Sternsdorf T, Phan VT, Maunakea ML, Ocampo CB, Sohal J, 4 Cools J, DeAngelo DJ, Gotlib J, Stover EH, Legare RD, Cortes J et al. Silletto A et al. Forced retinoic acid receptor alpha homodimers A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 prime mice for APL-like leukemia. Cancer Cell 2006; 9: 81–94. genes as a therapeutic target of imatinib in idiopathic hypereosino- 11 Kwok C, Zeisig BB, Dong S, So CW. Forced homo-oligomerization philic syndrome. N Engl J Med 2003; 348: 1201–1214. of RARalpha leads to transformation of primary hematopoietic 5 Kaufmann I, Martin G, Friedlein A, Langen H, Keller W. Human cells. Cancer Cell 2006; 9: 95–108. Fip1 is a subunit of CPSF that binds to U-rich RNA elements and 12 Palaniswamy V, Moraes KC, Wilusz CJ, Wilusz J. Nucleophosmin stimulates poly(A) polymerase. EMBO J 2004; 23: 616–626. is selectively deposited on mRNA during polyadenylation. Nat 6 Sainty D, Liso V, Cantu-Rajnoldi A, Head D, Mozziconacci MJ, Struct Mol Biol 2006; 13: 429–435. Arnoulet C et al. A new morphologic classification system for acute 13 Rego EM, Ruggero D, Tribioli C, Cattoretti G, Kogan S, Redner RL promyelocytic leukemia distinguishes cases with underlying PLZF/ et al.
    [Show full text]
  • Cldn19 Clic2 Clmp Cln3
    NewbornDx™ Advanced Sequencing Evaluation When time to diagnosis matters, the NewbornDx™ Advanced Sequencing Evaluation from Athena Diagnostics delivers rapid, 5- to 7-day results on a targeted 1,722-genes. A2ML1 ALAD ATM CAV1 CLDN19 CTNS DOCK7 ETFB FOXC2 GLUL HOXC13 JAK3 AAAS ALAS2 ATP1A2 CBL CLIC2 CTRC DOCK8 ETFDH FOXE1 GLYCTK HOXD13 JUP AARS2 ALDH18A1 ATP1A3 CBS CLMP CTSA DOK7 ETHE1 FOXE3 GM2A HPD KANK1 AASS ALDH1A2 ATP2B3 CC2D2A CLN3 CTSD DOLK EVC FOXF1 GMPPA HPGD K ANSL1 ABAT ALDH3A2 ATP5A1 CCDC103 CLN5 CTSK DPAGT1 EVC2 FOXG1 GMPPB HPRT1 KAT6B ABCA12 ALDH4A1 ATP5E CCDC114 CLN6 CUBN DPM1 EXOC4 FOXH1 GNA11 HPSE2 KCNA2 ABCA3 ALDH5A1 ATP6AP2 CCDC151 CLN8 CUL4B DPM2 EXOSC3 FOXI1 GNAI3 HRAS KCNB1 ABCA4 ALDH7A1 ATP6V0A2 CCDC22 CLP1 CUL7 DPM3 EXPH5 FOXL2 GNAO1 HSD17B10 KCND2 ABCB11 ALDOA ATP6V1B1 CCDC39 CLPB CXCR4 DPP6 EYA1 FOXP1 GNAS HSD17B4 KCNE1 ABCB4 ALDOB ATP7A CCDC40 CLPP CYB5R3 DPYD EZH2 FOXP2 GNE HSD3B2 KCNE2 ABCB6 ALG1 ATP8A2 CCDC65 CNNM2 CYC1 DPYS F10 FOXP3 GNMT HSD3B7 KCNH2 ABCB7 ALG11 ATP8B1 CCDC78 CNTN1 CYP11B1 DRC1 F11 FOXRED1 GNPAT HSPD1 KCNH5 ABCC2 ALG12 ATPAF2 CCDC8 CNTNAP1 CYP11B2 DSC2 F13A1 FRAS1 GNPTAB HSPG2 KCNJ10 ABCC8 ALG13 ATR CCDC88C CNTNAP2 CYP17A1 DSG1 F13B FREM1 GNPTG HUWE1 KCNJ11 ABCC9 ALG14 ATRX CCND2 COA5 CYP1B1 DSP F2 FREM2 GNS HYDIN KCNJ13 ABCD3 ALG2 AUH CCNO COG1 CYP24A1 DST F5 FRMD7 GORAB HYLS1 KCNJ2 ABCD4 ALG3 B3GALNT2 CCS COG4 CYP26C1 DSTYK F7 FTCD GP1BA IBA57 KCNJ5 ABHD5 ALG6 B3GAT3 CCT5 COG5 CYP27A1 DTNA F8 FTO GP1BB ICK KCNJ8 ACAD8 ALG8 B3GLCT CD151 COG6 CYP27B1 DUOX2 F9 FUCA1 GP6 ICOS KCNK3 ACAD9 ALG9
    [Show full text]
  • Synapsin I in PC1 2 Cells. I. Characterization of the Phosphoprotein and Effect of Chronic NGF Treatment
    The Journal of Neuroscience, May 1987, 7(5): 1294-l 299 Synapsin I in PC1 2 Cells. I. Characterization of the Phosphoprotein and Effect of Chronic NGF Treatment Carmelo Romano, Robert A. Nichols, Paul Greengard, and Lloyd A. Greene Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York 10021; and Department of Pharmacology, New York University School of Medicine, New York, New York 10016 PC1 2 cells contain a synapsin l-like molecule. Several serum Adrenal chromaffin cells and sympathetic neurons share a and monoclonal antibodies raised against bovine brain syn- neural crest origin and many other similarities (Coupland, 1965; apsin I bind to and precipitate this molecule, demonstrating Weston, 1970). Nevertheless, normal rat adrenal chromaffin immunochemical similarity between the brain and PC12 cells do not, whereassympathetic neurons do, contain synapsin species. PC12 synapsin I, like brain synapsin I, is a phos- I (DeCamilli et al., 1979; Fried et al., 1982). To better under- phoprotein: It is phosphorylated in intact cells and, when stand the developmental regulation of synapsin I, it was there- partially purified, serves as a substrate for several synapsin fore of interest to study synapsin I in PC 12 cells and its possible I kinases. PC1 2 cell synapsin I is structurally similar to brain alteration upon treatment of the cells with NGF. This paper synapsin I as shown by peptide mapping of %-methionine- characterizes the synapsin I present in PC12 cells and demon- and 32P-phosphate-labeled molecules from the 2 sources. strates effects of long-term NGF treatment of the cells on the Chronic NGF treatment of the cells induces a significant phosphoprotein.The accompanyingpaper (Roman0 et al., 1987) increase in the amount of synapsin I relative to total cell demonstratesthat short-term NGF treatment of PC12 cells re- protein, measured either by immunolabeling or incorporation sults in the phosphorylation of synapsin I at a novel site.
    [Show full text]
  • Genome-Wide Alterations in Gene Methylation by the BRAF V600E Mutation in Papillary Thyroid Cancer Cells
    Endocrine-Related Cancer (2011) 18 687–697 Genome-wide alterations in gene methylation by the BRAF V600E mutation in papillary thyroid cancer cells Peng Hou, Dingxie Liu and Mingzhao Xing Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology and Metabolism, The Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 333, Baltimore, Maryland 21287, USA (Correspondence should be addressed to M Xing; Email: [email protected]) Abstract The BRAF V600E mutation plays an important role in the tumorigenesis of papillary thyroid cancer (PTC). To explore an epigenetic mechanism involved in this process, we performed a genome- wide DNA methylation analysis using a methylated CpG island amplification (MCA)/CpG island microarray system to examine gene methylation alterations after shRNA knockdown of BRAF V600E in thyroid cancer cells. Our results revealed numerous methylation targets of BRAF V600E mutation with a large cohort of hyper- or hypo-methylated genes in thyroid cancer cells, which are known to have important metabolic and cellular functions. As hypomethylation of numerous genes by BRAF V600E was particularly a striking finding, we took a further step to examine the selected 59 genes that became hypermethylated in both cell lines upon BRAF V600E knockdown and found them to be mostly correspondingly under-expressed (i.e. they were normally maintained hypomethylated and over-expressed by BRAF V600E in thyroid cancer cells). We confirmed the methylation status of selected genes revealed on MCA/CpG microarray analysis by performing methylation-specific PCR. To provide proof of concept that some of the genes uncovered here may play a direct oncogenic role, we selected six of them to perform shRNA knockdown and examined its effect on cellular functions.
    [Show full text]
  • Figure S1. HAEC ROS Production and ML090 NOX5-Inhibition
    Figure S1. HAEC ROS production and ML090 NOX5-inhibition. (a) Extracellular H2O2 production in HAEC treated with ML090 at different concentrations and 24 h after being infected with GFP and NOX5-β adenoviruses (MOI 100). **p< 0.01, and ****p< 0.0001 vs control NOX5-β-infected cells (ML090, 0 nM). Results expressed as mean ± SEM. Fold increase vs GFP-infected cells with 0 nM of ML090. n= 6. (b) NOX5-β overexpression and DHE oxidation in HAEC. Representative images from three experiments are shown. Intracellular superoxide anion production of HAEC 24 h after infection with GFP and NOX5-β adenoviruses at different MOIs treated or not with ML090 (10 nM). MOI: Multiplicity of infection. Figure S2. Ontology analysis of HAEC infected with NOX5-β. Ontology analysis shows that the response to unfolded protein is the most relevant. Figure S3. UPR mRNA expression in heart of infarcted transgenic mice. n= 12-13. Results expressed as mean ± SEM. Table S1: Altered gene expression due to NOX5-β expression at 12 h (bold, highlighted in yellow). N12hvsG12h N18hvsG18h N24hvsG24h GeneName GeneDescription TranscriptID logFC p-value logFC p-value logFC p-value family with sequence similarity NM_052966 1.45 1.20E-17 2.44 3.27E-19 2.96 6.24E-21 FAM129A 129. member A DnaJ (Hsp40) homolog. NM_001130182 2.19 9.83E-20 2.94 2.90E-19 3.01 1.68E-19 DNAJA4 subfamily A. member 4 phorbol-12-myristate-13-acetate- NM_021127 0.93 1.84E-12 2.41 1.32E-17 2.69 1.43E-18 PMAIP1 induced protein 1 E2F7 E2F transcription factor 7 NM_203394 0.71 8.35E-11 2.20 2.21E-17 2.48 1.84E-18 DnaJ (Hsp40) homolog.
    [Show full text]
  • Agrin Controls Synaptic Differentiation in Hippocampal Neurons
    The Journal of Neuroscience, December 15, 2000, 20(24):9086–9095 Agrin Controls Synaptic Differentiation in Hippocampal Neurons Christian M. Bo¨ se,1 Dike Qiu,1 Andrea Bergamaschi,3 Biagio Gravante,3 Mario Bossi,2 Antonello Villa,2 Fabio Rupp,1 and Antonio Malgaroli3 1Department of Neuroscience, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, 2Microscopy and Image Analysis, University of Milan School of Medicine, and 3Department of Biological and Technological Research, Scientific Institute San Raffaele, 20123 Milano, Italy Agrin controls the formation of the neuromuscular junction. agrin and could not be detected in cultures from agrin-deficient Whether it regulates the differentiation of other types of synapses animals. Interestingly, the few synapses formed in reduced agrin remains unclear. Therefore, we have studied the role of agrin in conditions displayed diminished vesicular turnover, despite a cultured hippocampal neurons. Synaptogenesis was severely normal appearance at the EM level. Thus, our results demon- compromised when agrin expression or function was sup- strate the necessity of agrin for synaptogenesis in hippocampal pressed by antisense oligonucleotides and specific antibodies. neurons. The effects of antisense oligonucleotides were found to be highly Key words: agrin; synaptogenesis; synapses; primary hip- specific because they were reversed by adding recombinant pocampal neurons; agrin antibodies; antisense oligonucleotides The fidelity of neural transmission depends on interactions of a can be detected in CNS neurons, where the temporal pattern of large number of presynaptic and postsynaptic molecules. Despite expression of agrin parallels synaptogenesis (Hoch et al., 1993; the importance of these processes for neural communication, the O’Connor et al., 1994; Stone and Nikolics, 1995; N.
    [Show full text]
  • Genetic Testing Policy Number: PG0041 ADVANTAGE | ELITE | HMO Last Review: 04/11/2021
    Genetic Testing Policy Number: PG0041 ADVANTAGE | ELITE | HMO Last Review: 04/11/2021 INDIVIDUAL MARKETPLACE | PROMEDICA MEDICARE PLAN | PPO GUIDELINES This policy does not certify benefits or authorization of benefits, which is designated by each individual policyholder terms, conditions, exclusions and limitations contract. It does not constitute a contract or guarantee regarding coverage or reimbursement/payment. Paramount applies coding edits to all medical claims through coding logic software to evaluate the accuracy and adherence to accepted national standards. This medical policy is solely for guiding medical necessity and explaining correct procedure reporting used to assist in making coverage decisions and administering benefits. SCOPE X Professional X Facility DESCRIPTION A genetic test is the analysis of human DNA, RNA, chromosomes, proteins, or certain metabolites in order to detect alterations related to a heritable or acquired disorder. This can be accomplished by directly examining the DNA or RNA that makes up a gene (direct testing), looking at markers co-inherited with a disease-causing gene (linkage testing), assaying certain metabolites (biochemical testing), or examining the chromosomes (cytogenetic testing). Clinical genetic tests are those in which specimens are examined and results reported to the provider or patient for the purpose of diagnosis, prevention or treatment in the care of individual patients. Genetic testing is performed for a variety of intended uses: Diagnostic testing (to diagnose disease) Predictive
    [Show full text]
  • Supplementary Table 1: List of the 316 Genes Regulated During Hyperglycemic Euinsulinemic Clamp in Skeletal Muscle
    Supplementary Table 1: List of the 316 genes regulated during hyperglycemic euinsulinemic clamp in skeletal muscle. UGCluster Name Symbol Fold Change Cytoband Response to stress Hs.517581 Heme oxygenase (decycling) 1 HMOX1 3.80 22q12 Hs.374950 Metallothionein 1X MT1X 2.20 16q13 Hs.460867 Metallothionein 1B (functional) MT1B 1.70 16q13 Hs.148778 Oxidation resistance 1 OXR1 1.60 8q23 Hs.513626 Metallothionein 1F (functional) MT1F 1.47 16q13 Hs.534330 Metallothionein 2A MT2A 1.45 16q13 Hs.438462 Metallothionein 1H MT1H 1.42 16q13 Hs.523836 Glutathione S-transferase pi GSTP1 -1.74 11q13 Hs.459952 Stannin SNN -1.92 16p13 Immune response, cytokines & related Hs.478275 TNF (ligand) superfamily, member 10 (TRAIL) TNFSF10 1.58 3q26 Hs.278573 CD59 antigen p18-20 (protectin) CD59 1.49 11p13 Hs.534847 Complement component 4B, telomeric C4A 1.47 6p21.3 Hs.535668 Immunoglobulin lambda variable 6-57 IGLV6-57 1.40 22q11.2 Hs.529846 Calcium modulating ligand CAMLG -1.40 5q23 Hs.193516 B-cell CLL/lymphoma 10 BCL10 -1.40 1p22 Hs.840 Indoleamine-pyrrole 2,3 dioxygenase INDO -1.40 8p12-p11 Hs.201083 Mal, T-cell differentiation protein 2 MAL2 -1.44 Hs.522805 CD99 antigen-like 2 CD99L2 -1.45 Xq28 Hs.50002 Chemokine (C-C motif) ligand 19 CCL19 -1.45 9p13 Hs.350268 Interferon regulatory factor 2 binding protein 2 IRF2BP2 -1.47 1q42.3 Hs.567249 Contactin 1 CNTN1 -1.47 12q11-q12 Hs.132807 MHC class I mRNA fragment 3.8-1 3.8-1 -1.48 6p21.3 Hs.416925 Carcinoembryonic antigen-related cell adhesion molecule 19 CEACAM19 -1.49 19q13.31 Hs.89546 Selectin E (endothelial
    [Show full text]