Anti-POGZ Antibody, Rabbit Polyclonal

Total Page:16

File Type:pdf, Size:1020Kb

Anti-POGZ Antibody, Rabbit Polyclonal Anti-POGZ antibody, rabbit polyclonal 70-112 100 μl POGZ (pogo transposable element-derived protein with zinc finger domain; 1,410 aa, 155 kDa) plays a role in mitotic cell cycle progression and is involved in kinetochore assembly and mitotic sister chromatid cohesion. Probably through its association with CBX5 (HP1 alpha) it plays a role in mitotic chromosome segregation by regulating aurora kinase B/AURKB activation and AURKB and CBX5 dissociation from chromosome arms. Applications 1. Western Blotting (1/1,000 dilution) 2. Immunofluorescece staining (1/500 dilution) 3. Immunoprecipitation (assay dependent) Not tested for other applications Immunogen; Recombinant protein expressing 1-562 amino acids of human POGZ. Reactivity; Reacts specifically with human and mouse POGZ. Not tested in other species Form; Rabbit antiserum. No azide added. Sterilized by filtration through 0.22 μm membrane. Storage; Sent at 4℃ or -20℃. Store at -20℃. Avoid repeated freeze-thaw cycle. Data links; Entrez Gene: 23126 Human SwissProt: Q7Z3K3 Human Reference; This antibody was used in the following article. Nozawa R.S et al (2010) Human POGZ modulates dissociation of HP1alpha from mitotic chromosome arms through Aurora B activation. Nat. Cell Biol. 12:719-727 PubMed: 20562864 To be continued BioAcademia,Inc. Tel. 81-6-6877-2335 Fax. 81-6-6877-2336 [email protected] http://www.bioacademia.co.jp/en/ Fig.1 Western blot of POGZ Lane 1: Crude extract of HeLa cells transfected with control siRNA Lane 2: Crude extract of HeLa cells transfected with POGZ specific siRNA The antibody was used at 1/1,000 dilution. Specificity of the antibody to POGZ is confirmed by the disappearance of the POGZ band in the cells treated with the POGZ specific siRNA (lane 2) Fig.2 Immuno-fluorescence staining of POGZ. Paraformaldehyde fixed HeLa cells were stained with Hoechst 33342 and anti-POGZ antibody. The antibody was used at 1/500 dilution. Scale bar, 10 μm Fig. 3 Immuno-fluorescence staining of POGD on chromosomes. Chromosome spreads were prepared from HeLa cells and stained with Hoechst 33342 and anti-POGZ antibody. The antibody was used at 1/500 dilution. Scale bar, 10 μm BioAcademia,Inc. Tel. 81-6-6877-2335 Fax. 81-6-6877-2336 [email protected] http://www.bioacademia.co.jp/en/ .
Recommended publications
  • Multiple Cellular Proteins Interact with LEDGF/P75 Through a Conserved Unstructured Consensus Motif
    ARTICLE Received 19 Jan 2015 | Accepted 1 Jul 2015 | Published 6 Aug 2015 DOI: 10.1038/ncomms8968 Multiple cellular proteins interact with LEDGF/p75 through a conserved unstructured consensus motif Petr Tesina1,2,3,*, Katerˇina Cˇerma´kova´4,*, Magdalena Horˇejsˇ´ı3, Katerˇina Procha´zkova´1, Milan Fa´bry3, Subhalakshmi Sharma4, Frauke Christ4, Jonas Demeulemeester4, Zeger Debyser4, Jan De Rijck4,**, Va´clav Veverka1,** & Pavlı´na Rˇeza´cˇova´1,3,** Lens epithelium-derived growth factor (LEDGF/p75) is an epigenetic reader and attractive therapeutic target involved in HIV integration and the development of mixed lineage leukaemia (MLL1) fusion-driven leukaemia. Besides HIV integrase and the MLL1-menin complex, LEDGF/p75 interacts with various cellular proteins via its integrase binding domain (IBD). Here we present structural characterization of IBD interactions with transcriptional repressor JPO2 and domesticated transposase PogZ, and show that the PogZ interaction is nearly identical to the interaction of LEDGF/p75 with MLL1. The interaction with the IBD is maintained by an intrinsically disordered IBD-binding motif (IBM) common to all known cellular partners of LEDGF/p75. In addition, based on IBM conservation, we identify and validate IWS1 as a novel LEDGF/p75 interaction partner. Our results also reveal how HIV integrase efficiently displaces cellular binding partners from LEDGF/p75. Finally, the similar binding modes of LEDGF/p75 interaction partners represent a new challenge for the development of selective interaction inhibitors. 1 Institute of Organic Chemistry and Biochemistry of the ASCR, v.v.i., Flemingovo nam. 2, 166 10 Prague, Czech Republic. 2 Department of Genetics and Microbiology, Faculty of Science, Charles University in Prague, Vinicna 5, 128 44 Prague, Czech Republic.
    [Show full text]
  • Genetic and Genomic Analysis of Hyperlipidemia, Obesity and Diabetes Using (C57BL/6J × TALLYHO/Jngj) F2 Mice
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Nutrition Publications and Other Works Nutrition 12-19-2010 Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P. Stewart Marshall University Hyoung Y. Kim University of Tennessee - Knoxville, [email protected] Arnold M. Saxton University of Tennessee - Knoxville, [email protected] Jung H. Kim Marshall University Follow this and additional works at: https://trace.tennessee.edu/utk_nutrpubs Part of the Animal Sciences Commons, and the Nutrition Commons Recommended Citation BMC Genomics 2010, 11:713 doi:10.1186/1471-2164-11-713 This Article is brought to you for free and open access by the Nutrition at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Nutrition Publications and Other Works by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. Stewart et al. BMC Genomics 2010, 11:713 http://www.biomedcentral.com/1471-2164/11/713 RESEARCH ARTICLE Open Access Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P Stewart1, Hyoung Yon Kim2, Arnold M Saxton3, Jung Han Kim1* Abstract Background: Type 2 diabetes (T2D) is the most common form of diabetes in humans and is closely associated with dyslipidemia and obesity that magnifies the mortality and morbidity related to T2D. The genetic contribution to human T2D and related metabolic disorders is evident, and mostly follows polygenic inheritance. The TALLYHO/ JngJ (TH) mice are a polygenic model for T2D characterized by obesity, hyperinsulinemia, impaired glucose uptake and tolerance, hyperlipidemia, and hyperglycemia.
    [Show full text]
  • De Novo POGZ Mutations in Sporadic
    Matsumura et al. Journal of Molecular Psychiatry (2016) 4:1 DOI 10.1186/s40303-016-0016-x SHORT REPORT Open Access De novo POGZ mutations in sporadic autism disrupt the DNA-binding activity of POGZ Kensuke Matsumura1, Takanobu Nakazawa2*, Kazuki Nagayasu2, Nanaka Gotoda-Nishimura1, Atsushi Kasai1, Atsuko Hayata-Takano1, Norihito Shintani1, Hidenaga Yamamori3, Yuka Yasuda3, Ryota Hashimoto3,4 and Hitoshi Hashimoto1,2,4 Abstract Background: A spontaneous de novo mutation is a new mutation appeared in a child that neither the parent carries. Recent studies suggest that recurrent de novo loss-of-function mutations identified in patients with sporadic autism spectrum disorder (ASD) play a key role in the etiology of the disorder. POGZ is one of the most recurrently mutated genes in ASD patients. Our laboratory and other groups have recently found that POGZ has at least 18 independent de novo possible loss-of-function mutations. Despite the apparent importance, these mutations have never previously been assessed via functional analysis. Methods: Using wild-type, the Q1042R-mutated, and R1008X-mutated POGZ, we performed DNA-binding experiments for proteins that used the CENP-B box sequence in vitro. Data were statistically analyzed by one-way ANOVA followed by Tukey-Kramer post hoc tests. Results: This study reveals that ASD-associated de novo mutations (Q1042R and R1008X) in the POGZ disrupt its DNA-binding activity. Conclusions: Here, we report the first functional characterization of de novo POGZ mutations identified in sporadic ASD cases. These findings provide important insights into the cellular basis of ASD. Keywords: Autism spectrum disorder, Recurrent mutation, De novo mutation, POGZ, DNA-binding activity Background including CHD8, ARID1B, SYNGAP1, DYRK1A, SCN2A, The genetic etiology of autism spectrum disorder (ASD) ANK2, ADNP, DSCAM, CHD2, KDM5B, SUV420H1, remains poorly understood.
    [Show full text]
  • POGZ Gene Pogo Transposable Element Derived with ZNF Domain
    POGZ gene pogo transposable element derived with ZNF domain Normal Function The POGZ gene provides instructions for making a protein that is found in the cell nucleus. The POGZ protein is part of a group known as zinc finger proteins, which contain one or more short regions called zinc finger domains. These regions include a specific pattern of protein building blocks (amino acids) and one or more charged atoms of zinc (zinc ions). The folded configuration of the zinc finger domain stabilizes the protein and allows it to attach (bind) to other molecules. In the cell nucleus, the POGZ protein attaches (binds) to chromatin, which is the network of DNA and proteins that packages DNA into chromosomes. Binding of the POGZ protein is part of the process that changes the structure of chromatin (chromatin remodeling) to alter how tightly regions of DNA are packaged. Chromatin remodeling is one way gene activity (expression) is regulated; when DNA is tightly packed gene expression is lower than when DNA is loosely packed. Regulation of gene expression by the POGZ protein is thought to be important to brain development, but the specific function of POGZ in the brain is not well understood. Health Conditions Related to Genetic Changes White-Sutton syndrome At least 17 POGZ gene mutations have been found to cause White-Sutton syndrome. This disorder is characterized by intellectual disability, specific facial features, and other signs and symptoms affecting various parts of the body, particularly vision problems and gastrointestinal problems. Most affected individuals have features of autism spectrum disorder (ASD), a varied condition characterized by impaired social skills, communication problems, and repetitive behaviors.
    [Show full text]
  • Molecular Mechanism of LEDGF/P75 Dimerization
    Article Molecular Mechanism of LEDGF/p75 Dimerization Graphical Abstract Authors Vanda Lux, Tine Brouns, Katerina Cerma ´ kova´ , ..., Frauke Christ, Zeger Debyser, Va´ clav Veverka Correspondence [email protected] (Z.D.), [email protected] (V.V.) In Brief Dimerization is an important process regulating eukaryotic transcription. Lux et al. use a combination of biophysical and biochemical techniques to investigate the dimerization mechanism of LEDGF/p75, the H3K36 methylation reader, and its effect on molecular interactions with other proteins. Highlights d Full-length LEDGF/p75 forms dimers with a low micromolar KD d LEDGF/p75 residues 345–467 is the minimal stable dimerization domain d LEDGF dimer is stabilized by domain swapping and additional electrostatic stapling d LEDGF dimerization does not impair binding of interaction partners Lux et al., 2020, Structure 28, 1288–1299 December 1, 2020 ª 2020 Elsevier Ltd. https://doi.org/10.1016/j.str.2020.08.012 ll ll Article Molecular Mechanism of LEDGF/p75 Dimerization Vanda Lux,1,7 Tine Brouns,2,7 Katerina Cerma´ kova´ ,1,3 Pavel Srb,1 Milan Fa´ bry,4 Marcela Ma´ dlı´kova´ ,1 Magdalena Horejsı´,4 Zdenek Kukacka, 5 Petr Nova´ k,5 Michael Kugler,1 Jirı´ Brynda,1,4 Jan DeRijck,2 Frauke Christ,2 Zeger Debyser,2,* and Va´ clav Veverka1,6,8,* 1Structural Biology, Institute of Organic Chemistry and Biochemistry of the CAS, Prague 16000, Czech Republic 2Molecular Virology and Gene Therapy, KU Leuven, Molecular Virology and Gene Therapy, Leuven, 3000 Flanders, Belgium 3Department
    [Show full text]
  • Association of Polymorphisms in the LEDGF/P75 Gene (PSIP1) with Susceptibility to HIV-1 Infection and Disease Progression
    Association of polymorphisms in the LEDGF/p75 gene (PSIP1) with susceptibility to HIV-1 infection and disease progression Paradise Madlalaa,b, Rik Gijsbersc, Frauke Christc, Anneleen Hombrouckc, Lise Wernerd, Koleka Mlisanad, Ping Ane, Salim S. Abdool Karimd, Cheryl A. Winklere, Zeger Debyserc and Thumbi Ndung’ua,d Objective: LEDGF/p75, encoded by the PSIP1 gene, interacts with HIV-1 integrase and targets HIV-1 integration into active genes. We investigated the influence of poly- morphisms in PSIP1 on HIV-1 acquisition and disease progression in black South Africans. Methods: Integrase binding domain of LEDGF/p75 was sequenced in 126 participants. Four haplotype tagging SNPs rs2277191, rs1033056, rs12339417 and rs10283923 referred to as SNP1, SNP2, SNP3 and SNP4, respectively, and one exonic SNP rs61744944 (SNP5, Q472L) were genotyped in 195 HIV-1 seronegative, 52 primary and 403 chronically infected individuals using TaqMan assays. LEDGF/p75 expression was quantified by real-time RT-PCR. The impact of Q472L mutation on the interaction with HIV_1 IN was measured by AlphaScreen. Results: rs2277191 (SNP1) A was more frequent among seropositives (P ¼ 0.06, Fish- er’s exact test). Among individuals followed longitudinally SNP1A trended towards association with higher likelihood of HIV-1 acquisition [relative hazard (RH) ¼ 2.21, P ¼ 0.08; Cox model] and it was also associated with rapid disease progression (RH ¼ 5.98, P ¼ 0.04; Cox model) in the recently infected (primary infection) cohort. rs12339417 (SNP3)C was associated with slower decline of CD4þ T cells (P ¼ 0.02) and lower messenger RNA (mRNA) levels of LEDGF/p75 (P < 0.01).
    [Show full text]
  • Peptide-Binding Repertoires HLA-A*02 Have Overlapping Patr
    Although Divergent in Residues of the Peptide Binding Site, Conserved Chimpanzee Patr-AL and Polymorphic Human HLA-A*02 Have Overlapping This information is current as Peptide-Binding Repertoires of September 26, 2021. Michael Gleimer, Angela R. Wahl, Heather D. Hickman, Laurent Abi-Rached, Paul J. Norman, Lisbeth A. Guethlein, John A. Hammond, Monia Draghi, Erin J. Adams, Sean Juo, Roxana Jalili, Baback Gharizadeh, Mostafa Ronaghi, K. Downloaded from Christopher Garcia, William H. Hildebrand and Peter Parham J Immunol 2011; 186:1575-1588; Prepublished online 5 January 2011; doi: 10.4049/jimmunol.1002990 http://www.jimmunol.org/ http://www.jimmunol.org/content/186/3/1575 Supplementary http://www.jimmunol.org/content/suppl/2011/01/05/jimmunol.100299 Material 0.DC1 References This article cites 87 articles, 24 of which you can access for free at: by guest on September 26, 2021 http://www.jimmunol.org/content/186/3/1575.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2011 by The American Association of Immunologists, Inc.
    [Show full text]
  • TBR1 Regulates Autism Risk Genes in the Developing Neocortex
    Downloaded from genome.cshlp.org on October 6, 2021 - Published by Cold Spring Harbor Laboratory Press Research TBR1 regulates autism risk genes in the developing neocortex James H. Notwell,1 Whitney E. Heavner,2,3 Siavash Fazel Darbandi,4 Sol Katzman,5 William L. McKenna,6 Christian F. Ortiz-Londono,6 David Tastad,6 Matthew J. Eckler,6 John L.R. Rubenstein,4 Susan K. McConnell,3 Bin Chen,6 and Gill Bejerano1,2,7 1Department of Computer Science, 2Department of Developmental Biology, 3Department of Biology, Stanford University, Stanford, California 94305, USA; 4Department of Psychiatry, University of California, San Francisco, San Francisco, California, 94143 USA; 5Center for Biomolecular Science and Engineering, University of California, Santa Cruz, Santa Cruz, California 95064 USA; 6Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California 95064, USA; 7Division of Medical Genetics, Department of Pediatrics, Stanford University, Stanford, California 94305, USA Exome sequencing studies have identified multiple genes harboring de novo loss-of-function (LoF) variants in individuals with autism spectrum disorders (ASD), including TBR1, a master regulator of cortical development. We performed ChIP- seq for TBR1 during mouse cortical neurogenesis and show that TBR1-bound regions are enriched adjacent to ASD genes. ASD genes were also enriched among genes that are differentially expressed in Tbr1 knockouts, which together with the ChIP-seq data, suggests direct transcriptional regulation. Of the nine ASD genes examined, seven were misexpressed in the cortices of Tbr1 knockout mice, including six with increased expression in the deep cortical layers. ASD genes with adjacent cortical TBR1 ChIP-seq peaks also showed unusually low levels of LoF mutations in a reference human population and among Icelanders.
    [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]
  • Twenty Years of Research on the DFS70/LEDGF Autoantibody
    Ortiz‑Hernandez et al. Autoimmun Highlights (2020) 11:3 https://doi.org/10.1186/s13317‑020‑0126‑4 Autoimmunity Highlights REVIEW Open Access Twenty years of research on the DFS70/ LEDGF autoantibody‑autoantigen system: many lessons learned but still many questions Greisha L. Ortiz‑Hernandez1,2, Evelyn S. Sanchez‑Hernandez1,2 and Carlos A. Casiano1,2,3* Abstract The discovery and initial characterization 20 years ago of antinuclear autoantibodies (ANAs) presenting a dense fne speckled (DFS) nuclear pattern with strong staining of mitotic chromosomes, detected by indirect immunofuores‑ cence assay in HEp‑2 cells (HEp‑2 IIFA test), has transformed our view on ANAs. Traditionally, ANAs have been consid‑ ered as reporters of abnormal immunological events associated with the onset and progression of systemic autoim‑ mune rheumatic diseases (SARD), also called ANA‑associated rheumatic diseases (AARD), as well as clinical biomarkers for the diferential diagnosis of these diseases. However, based on our current knowledge, it is not apparent that autoantibodies presenting the DFS IIF pattern fall into these categories. These antibodies invariably target a chroma‑ tin‑associated protein designated as dense fne speckled protein of 70 kD (DFS70), also known as lens epithelium‑ derived growth factor protein of 75 kD (LEDGF/p75) and PC4 and SFRS1 Interacting protein 1 (PSIP1). This multi‑func‑ tional protein, hereafter referred to as DFS70/LEDGF, plays important roles in the formation of transcription complexes in active chromatin, transcriptional activation of specifc genes, regulation of mRNA splicing, DNA repair, and cellular survival against stress. Due to its multiple functions, it has emerged as a key protein contributing to several human pathologies, including acquired immunodefciency syndrome (AIDS), leukemia, cancer, ocular diseases, and Rett syn‑ drome.
    [Show full text]
  • Recurrent De Novo Mutations in Neurodevelopmental Disorders: Properties and Clinical Implications Amy B
    Wilfert et al. Genome Medicine (2017) 9:101 DOI 10.1186/s13073-017-0498-x REVIEW Open Access Recurrent de novo mutations in neurodevelopmental disorders: properties and clinical implications Amy B. Wilfert1†, Arvis Sulovari1†, Tychele N. Turner1, Bradley P. Coe1 and Evan E. Eichler1,2* Abstract Next-generation sequencing (NGS) is now more accessible to clinicians and researchers. As a result, our understanding of the genetics of neurodevelopmental disorders (NDDs) has rapidly advanced over the past few years. NGS has led to the discovery of new NDD genes with an excess of recurrent de novo mutations (DNMs) when compared to controls. Development of large-scale databases of normal and disease variation has given rise to metrics exploring the relative tolerance of individual genes to human mutation. Genetic etiology and diagnosis rates have improved, which have led to the discovery of new pathways and tissue types relevant to NDDs. In this review, we highlight several key findings based on the discovery of recurrent DNMs ranging from copy number variants to point mutations. We explore biases and patterns of DNM enrichment and the role of mosaicism and secondary mutations in variable expressivity. We discuss the benefit of whole-genome sequencing (WGS) over whole-exome sequencing (WES) to understand more complex, multifactorial cases of NDD and explain how this improved understanding aids diagnosis and management of these disorders. Comprehensive assessment of the DNM landscape across the genome using WGS and other technologies will lead to the development of novel functional and bioinformatics approaches to interpret DNMs and drive new insights into NDD biology.
    [Show full text]
  • Pogz Deficiency Leads to Abnormal Behavior, Transcription Dysregulation And
    bioRxiv preprint doi: https://doi.org/10.1101/437442; this version posted October 8, 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. Pogz deficiency leads to abnormal behavior, transcription dysregulation and impaired cerebellar physiology Reut Suliman1,*, Ben Title2,*, Yahel Cohen1, Maayan Tal1, Nitzan Tal1, Bjorg Gudmundsdottir3, Kristbjorn O. Gudmundsson3, Jonathan R Keller3,4, Guo-Jen Huang5, Yosef Yarom2, a and Sagiv Shifman1, b 1 Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel 2 Department of Neurobiology, The Institute of Life Sciences and Edmond & Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem, Israel 3 Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute at Frederick, Bldg. 560/12-70, 1050 Boyles Street, Frederick, MD 21702, USA 4 Basic Research Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Bldg. 560/32-31D, 1050 Boyles Street, Frederick, MD 21702, USA 5 Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan * Equal contribution Correspondence: a Yosef Yarom, Department of Neurobiology, The Institute of life sciences, The Hebrew University of Jerusalem, Edmond J. Safra campus,, Jerusalem 91904, Israel Email: [email protected], Phone: +972-2-6585172, Fax: 972-2-6586926 b Sagiv Shifman, Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J.
    [Show full text]