The Cytoskeletal Protein RHAMM and ERK1/2 Activity Maintain the Pluripotency of Murine Embryonic Stem Cells

Total Page:16

File Type:pdf, Size:1020Kb

The Cytoskeletal Protein RHAMM and ERK1/2 Activity Maintain the Pluripotency of Murine Embryonic Stem Cells The Cytoskeletal Protein RHAMM and ERK1/2 Activity Maintain the Pluripotency of Murine Embryonic Stem Cells Jihong Jiang, Pooja Mohan, Christopher A. Maxwell* Department of Pediatrics, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada Abstract Receptor for hyaluronan mediated motility (RHAMM, encoded by HMMR) may be a cell-surface receptor for hyaluronan that regulates embryonic stem cell pluripotency and differentiation, however, a precise mechanism for its action is not known. We examined murine embryonic stem cells with and without hemizygous genomic mutation of Hmmr/RHAMM, but we were not able to find RHAMM on the cell-surface. Rather, RHAMM localized to the microtubule cytoskeleton and along mitotic spindles. Genomic loss of Hmmr/RHAMM did not alter cell cycle progression but augmented differentiation and attenuated pluripotency in murine embryonic stem cells. Through a candidate screen of small-molecule kinase inhibitors, we identified ERK1/2 and aurora kinase A as barrier kinases whose inhibition was sufficient to rescue pluripotency in RHAMM+/- murine embryonic stem cells. Thus, RHAMM is not found on the cell-surface of embryonic stem cells, but it is required to maintain pluripotency and its dominant mechanism of action is through the modulation of signal transduction pathways at microtubules. Citation: Jiang J, Mohan P, Maxwell CA (2013) The Cytoskeletal Protein RHAMM and ERK1/2 Activity Maintain the Pluripotency of Murine Embryonic Stem Cells. PLoS ONE 8(9): e73548. doi:10.1371/journal.pone.0073548 Editor: Austin John Cooney, Baylor College of Medicine, United States of America Received April 17, 2013; Accepted July 19, 2013; Published September 3, 2013 Copyright: © 2013 Jiang et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Funding: These studies were supported by a Child and Youth Health, New Investigator Award from the SickKids Foundation and the Institute of Human Development Child and Youth Health-CIHR (Grant No. NI11- 005), and by a Pediatric Brain Tumor grant from the Michael Cuccione Foundation for Childhood Cancer Research. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Competing interests: The authors have declared that no competing interests exist. * E-mail: [email protected] Introduction Originally identified as a peptide in the supernatants from proliferative fibroblasts [8], RHAMM may be passively released Hyaluronan (HA) is an extracellular polysaccharide, and HA- to the extracellular space through the cell death that rich hydrogels maintain human embryonic stem (ES) cells in accompanies pathological states, such as cancer and their undifferentiated state [1]. Extracellular receptors for HA inflammation [9]. However, RHAMM is an intracellular protein are likely responsible for the transmission of intracellular that associates with both microtubules and actin microfilaments signals that maintain ES cell pluripotency. Indeed, CD44, the [10]. RHAMM forms a complex with the dynein molecular motor major cellular receptor for HA, is critical to the expansion, to localize to the centrosome [11] and enable mitotic spindle differentiation, and pluripotency of a wide range of stem cells, assembly [12–14], and RHAMM regulates signal transduction including cancer stem cells [2], neural [3], mesenchymal [4] at microtubules, including the aurora kinase A (AURKA) [14] and epidermal stem cells [5]. Moreover, human ES cells also and ERK1/2 [15] pathways. Thus, the putative physiological express receptor for hyaluronan mediated motility (RHAMM, role is not yet clear for RHAMM during the self-renewal of ES encoded by HMMR) [6], a putative and controversial receptor cells. for HA [7]. The expression of RHAMM is dramatically reduced Here, we examine the localization and action of RHAMM with differentiation in vitro, and the silencing of RHAMM with within mouse ES cells, which contain an insertional mutation in siRNA disrupts the self-renewal of human ES cells [6]. Thus, one allele of the Hmmr/RHAMM gene. We do not find that the extracellular localization of RHAMM, and its putative RHAMM is expressed on the cell-surface but rather that engagement of HA, may be a critical regulatory cue that RHAMM is a cytoskeletal and mitotic spindle protein in these decides the cellular fate of ES cells; such an action for RHAMM cells. The hemizygous genomic loss of Hmmr/RHAMM would be vital to multiple aspects of stem cell biology, including attenuates mouse ES cell pluripotency and we use a small- stem cell niches, tissue engineering, and biomaterials. molecule screen to discover that the phosphorylation of PLOS ONE | www.plosone.org 1 September 2013 | Volume 8 | Issue 9 | e73548 RHAMM and Pluripotency ERK1/2 and AURKA are elevated and these kinases act as 3VO4 and 50 mM NaF. Cell lysates were clarified by barriers to pluripotency in RHAMM+/- mouse ES cells. centrifugation at 16,000 X g for 10 min at 4oC, and protein concentration was determined by BCA protein assay kit Materials and Methods (Thermal Scientific). Cell lysates were mixed with SDS sample buffer, separated by SDS-PAGE, and blotted with the following Mouse ES cell Culture antibodies: anti-RHAMM (Epitomics), anti-Oct3/4 (StemCell RHAMM+/- mouse ES cell-lines (BB0166- MMRRC: 026467- Technologies), anti-NUMB (Abcam), anti-(p) ERK1/2 (Cell UCD; and XP0038- MMRRC: 028514-UCD) with mutational Signaling), anti-ERK (Cell Signaling), anti-AURKA (Abcam), inactivation of one allele for the Hmmr/RHAMM gene and the anti-(p) AURKA (Cell Signaling), and anti-Actin (Sigma). parental control RHAMM+/+ mouse ES cell-line (E14TG2a) were purchased from the International Gene Trap Consortium Fluorescent Activated Cell Sorting Analysis through a Mutant Mouse Regional Resource Center (University Mouse ES cells were harvested by trypsinization and of California, Davis). Briefly, mouse ES cells were maintained washed. For cell surface protein expression, cells were on mitomycin C-treated mouse embryonic fibroblast (MEF) incubated with the primary antibodies (30 min, 4oC), washed cells in medium consisting of high-glucose Dulbecco’s modified twice, and then incubated with PE-conjugated anti-mouse IgM Eagle’s medium (DMEM) supplemented with fetal bovine (BD pharmingen) or anti-rabbit Alexa-647 IgG (30 min, 4oC) serum (FBS) 16% (ES-qualified, Invitrogen), L-glutamine 2 mM, and washed twice. For intracellular protein expression, cells non-essential amino acids (NEAA) 0.1 mM, leukemia inhibitory were fixed and permeabilized with fresh 4% PFA (15 min, RT) factor (LIF) 1000 U/ml (Chemicon), and 2-mercaptoethanol 143 and methanol (10 mins, -20°C), or methanol alone, before µM. For feeder-free cultures, mouse ES cells were grown on immunostaining. The following primary antibodies were used in CellBind plates (Corning) in medium consisting of Glasgow this study: anti-SSEA (StemCell Technologies), anti-N terminal minimal essential medium (GMEM) (Sigma) containing FBS RHAMM (Epitomics), anti-C terminal RHAMM (Epitomics), anti- 15% (ES-qualified, Invitrogen), L-glutamine 2 mM, sodium TPX2 (Novus). For cell cycle analysis, cells were fixed with pyruvate 1 mM, NEAA 0.1 mM, LIF 1000 U/ml, and 2- 70% ethanol at -20°C overnight, and then stained with 60 µg/ml mercaptoethanol 66 µM. propidium iodide (Invitrogen) for 30 min. FACS analysis was performed using a FACSCalibure2 flow cytometer (BD Quantitative Reverse Transcriptase Polymerase Chain biosciences) and the CellQuest software. Reaction (RT-PCR) Total RNA was extracted from cells with a RNeasy mini kit Cell proliferation (Qiagen), treated with DNase I (Invitrogen), and converted to To measure the doubling time for mouse ES cell-lines, 105 cDNA with a high capacity cDNA RT kit (Applied Biosystems). cells were seeded in 24 well CellBind plates. Cell numbers Real-time quantitative RT-PCR were performed as described were counted 24, 48, 72 and 96 hours after plating. Doubling [16] using the primers listed in Table S1. time was calculated by the equation Yend = Ystart × 2(t/T), where T is the doubling time, Ystart is the starting number of Immunostaining ES cells (24 hours after plating) and Yend is the ending Cells were grown on coverslips coated with 0.1% gelatin, number of ES cells after a period of time t. fixed with 4% paraformaldehyde (PFA) for 15 minutes (min) at room temperature (RT). Cells were permeablized and blocked Alkaline Phosphatase (AP) Assays and Embryoid Body with 0.3% triton X-100, 10% normal donkey serum, 0.1% BSA Formation in PBS, and incubated with antibody listed in Table S2 at their Cells were stained with alkaline phosphatase staining kit II respective dilutions. For staining the antigen on the cell (Stemgent) following the manufacture’s instruction. Images surface, detergent was omitted from the buffer. were acquired with a Zeiss Axiovert 200 M inverted Following three washes in PBST, the following secondary microscope. Static suspension culture in knockout DMEM antibodies were applied for 1 hour at RT: anti-mouse (Invitrogen) containing 15% knockout serum replacement Alexa-488, anti-mouse Alexa-594, anti-mouse Alexa-647, anti- (Invitrogen) on ultra low attachment culture dishes was utilized rabbit Alexa-488, anti-rabbit Alexa-594, or anti-rabbit Alexa-647 to form embryoid bodies from Rhamm+/+ and Rhamm+/- ES
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Genome-Wide Transcriptional Sequencing Identifies Novel Mutations in Metabolic Genes in Human Hepatocellular Carcinoma DAOUD M
    CANCER GENOMICS & PROTEOMICS 11 : 1-12 (2014) Genome-wide Transcriptional Sequencing Identifies Novel Mutations in Metabolic Genes in Human Hepatocellular Carcinoma DAOUD M. MEERZAMAN 1,2 , CHUNHUA YAN 1, QING-RONG CHEN 1, MICHAEL N. EDMONSON 1, CARL F. SCHAEFER 1, ROBERT J. CLIFFORD 2, BARBARA K. DUNN 3, LI DONG 2, RICHARD P. FINNEY 1, CONSTANCE M. CULTRARO 2, YING HU1, ZHIHUI YANG 2, CU V. NGUYEN 1, JENNY M. KELLEY 2, SHUANG CAI 2, HONGEN ZHANG 2, JINGHUI ZHANG 1,4 , REBECCA WILSON 2, LAUREN MESSMER 2, YOUNG-HWA CHUNG 5, JEONG A. KIM 5, NEUNG HWA PARK 6, MYUNG-SOO LYU 6, IL HAN SONG 7, GEORGE KOMATSOULIS 1 and KENNETH H. BUETOW 1,2 1Center for Bioinformatics and Information Technology, National Cancer Institute, Rockville, MD, U.S.A.; 2Laboratory of Population Genetics, National Cancer Institute, National Cancer Institute, Bethesda, MD, U.S.A.; 3Basic Prevention Science Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, U.S.A; 4Department of Biotechnology/Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN, U.S.A.; 5Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea; 6Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan, Korea; 7Department of Internal Medicine, College of Medicine, Dankook University, Cheon-An, Korea Abstract . We report on next-generation transcriptome Worldwide, liver cancer is the fifth most common cancer and sequencing results of three human hepatocellular carcinoma the third most common cause of cancer-related mortality (1). tumor/tumor-adjacent pairs.
    [Show full text]
  • Analysis of the Indacaterol-Regulated Transcriptome in Human Airway
    Supplemental material to this article can be found at: http://jpet.aspetjournals.org/content/suppl/2018/04/13/jpet.118.249292.DC1 1521-0103/366/1/220–236$35.00 https://doi.org/10.1124/jpet.118.249292 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 366:220–236, July 2018 Copyright ª 2018 by The American Society for Pharmacology and Experimental Therapeutics Analysis of the Indacaterol-Regulated Transcriptome in Human Airway Epithelial Cells Implicates Gene Expression Changes in the s Adverse and Therapeutic Effects of b2-Adrenoceptor Agonists Dong Yan, Omar Hamed, Taruna Joshi,1 Mahmoud M. Mostafa, Kyla C. Jamieson, Radhika Joshi, Robert Newton, and Mark A. Giembycz Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada Received March 22, 2018; accepted April 11, 2018 Downloaded from ABSTRACT The contribution of gene expression changes to the adverse and activity, and positive regulation of neutrophil chemotaxis. The therapeutic effects of b2-adrenoceptor agonists in asthma was general enriched GO term extracellular space was also associ- investigated using human airway epithelial cells as a therapeu- ated with indacaterol-induced genes, and many of those, in- tically relevant target. Operational model-fitting established that cluding CRISPLD2, DMBT1, GAS1, and SOCS3, have putative jpet.aspetjournals.org the long-acting b2-adrenoceptor agonists (LABA) indacaterol, anti-inflammatory, antibacterial, and/or antiviral activity. Numer- salmeterol, formoterol, and picumeterol were full agonists on ous indacaterol-regulated genes were also induced or repressed BEAS-2B cells transfected with a cAMP-response element in BEAS-2B cells and human primary bronchial epithelial cells by reporter but differed in efficacy (indacaterol $ formoterol .
    [Show full text]
  • Hyaluronan Mediated Motility Receptor (HMMR) Encodes an Evolutionarily Conserved Homeostasis, Mitosis, and Meiosis Regulator Rather Than a Hyaluronan Receptor
    cells Review Hyaluronan Mediated Motility Receptor (HMMR) Encodes an Evolutionarily Conserved Homeostasis, Mitosis, and Meiosis Regulator Rather than a Hyaluronan Receptor 1 1 1, 1,2, Zhengcheng He , Lin Mei , Marisa Connell y and Christopher A. Maxwell * 1 Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; [email protected] (Z.H.); [email protected] (L.M.); [email protected] (M.C.) 2 Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital, Vancouver, BC V5Z 4H4, Canada * Correspondence: [email protected]; Tel.: +1-6048752000 (ext. 4691) Current position: Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, y NY 13210, USA. Received: 3 March 2020; Accepted: 25 March 2020; Published: 28 March 2020 Abstract: Hyaluronan is an extracellular matrix component that absorbs water in tissues and engages cell surface receptors, like Cluster of Differentiation 44 (CD44), to promote cellular growth and movement. Consequently, CD44 demarks stem cells in normal tissues and tumor-initiating cells isolated from neoplastic tissues. Hyaluronan mediated motility receptor (HMMR, also known as RHAMM) is another one of few defined hyaluronan receptors. HMMR is also associated with neoplastic processes and its role in cancer progression is often attributed to hyaluronan-mediated signaling. But, HMMR is an intracellular, microtubule-associated, spindle assembly factor that localizes protein complexes to augment the activities of mitotic kinases, like polo-like kinase 1 and Aurora kinase A, and control dynein and kinesin motor activities. Expression of HMMR is elevated in cells prior to and during mitosis and tissues with detectable HMMR expression tend to be highly proliferative, including neoplastic tissues.
    [Show full text]
  • Supplementary Material Contents
    Supplementary Material Contents Immune modulating proteins identified from exosomal samples.....................................................................2 Figure S1: Overlap between exosomal and soluble proteomes.................................................................................... 4 Bacterial strains:..............................................................................................................................................4 Figure S2: Variability between subjects of effects of exosomes on BL21-lux growth.................................................... 5 Figure S3: Early effects of exosomes on growth of BL21 E. coli .................................................................................... 5 Figure S4: Exosomal Lysis............................................................................................................................................ 6 Figure S5: Effect of pH on exosomal action.................................................................................................................. 7 Figure S6: Effect of exosomes on growth of UPEC (pH = 6.5) suspended in exosome-depleted urine supernatant ....... 8 Effective exosomal concentration....................................................................................................................8 Figure S7: Sample constitution for luminometry experiments..................................................................................... 8 Figure S8: Determining effective concentration .........................................................................................................
    [Show full text]
  • A Quantitative Chaperone Interaction Network Reveals the Architecture of Cellular Protein Homeostasis Pathways
    A Quantitative Chaperone Interaction Network Reveals the Architecture of Cellular Protein Homeostasis Pathways The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Taipale, Mikko et al. “A Quantitative Chaperone Interaction Network Reveals the Architecture of Cellular Protein Homeostasis Pathways.” Cell 158.2 (2014): 434–448. As Published http://dx.doi.org/10.1016/j.cell.2014.05.039 Publisher Elsevier Version Author's final manuscript Citable link http://hdl.handle.net/1721.1/105723 Terms of Use Creative Commons Attribution-NonCommercial-NoDerivs License Detailed Terms http://creativecommons.org/licenses/by-nc-nd/4.0/ NIH Public Access Author Manuscript Cell. Author manuscript; available in PMC 2015 July 17. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Cell. 2014 July 17; 158(2): 434–448. doi:10.1016/j.cell.2014.05.039. A quantitative chaperone interaction network reveals the architecture of cellular protein homeostasis pathways Mikko Taipale1, George Tucker2,3, Jian Peng2,3, Irina Krykbaeva1, Zhen-Yuan Lin4, Brett Larsen4, Hyungwon Choi5, Bonnie Berger2,3, Anne-Claude Gingras4,6, and Susan Lindquist1,7,8 1Whitehead Institute for Biomedical Research, Cambridge, MA 02114, USA 2Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 3Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 4Centre for Systems
    [Show full text]
  • Human Social Genomics in the Multi-Ethnic Study of Atherosclerosis
    Getting “Under the Skin”: Human Social Genomics in the Multi-Ethnic Study of Atherosclerosis by Kristen Monét Brown A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Epidemiological Science) in the University of Michigan 2017 Doctoral Committee: Professor Ana V. Diez-Roux, Co-Chair, Drexel University Professor Sharon R. Kardia, Co-Chair Professor Bhramar Mukherjee Assistant Professor Belinda Needham Assistant Professor Jennifer A. Smith © Kristen Monét Brown, 2017 [email protected] ORCID iD: 0000-0002-9955-0568 Dedication I dedicate this dissertation to my grandmother, Gertrude Delores Hampton. Nanny, no one wanted to see me become “Dr. Brown” more than you. I know that you are standing over the bannister of heaven smiling and beaming with pride. I love you more than my words could ever fully express. ii Acknowledgements First, I give honor to God, who is the head of my life. Truly, without Him, none of this would be possible. Countless times throughout this doctoral journey I have relied my favorite scripture, “And we know that all things work together for good, to them that love God, to them who are called according to His purpose (Romans 8:28).” Secondly, I acknowledge my parents, James and Marilyn Brown. From an early age, you two instilled in me the value of education and have been my biggest cheerleaders throughout my entire life. I thank you for your unconditional love, encouragement, sacrifices, and support. I would not be here today without you. I truly thank God that out of the all of the people in the world that He could have chosen to be my parents, that He chose the two of you.
    [Show full text]
  • Genetic Predictors of Hyperglycemia Due to Hydrochlorothiazide Therapy
    The Texas Medical Center Library DigitalCommons@TMC The University of Texas MD Anderson Cancer Center UTHealth Graduate School of The University of Texas MD Anderson Cancer Biomedical Sciences Dissertations and Theses Center UTHealth Graduate School of (Open Access) Biomedical Sciences 5-2012 GENETIC PREDICTORS OF HYPERGLYCEMIA DUE TO HYDROCHLOROTHIAZIDE THERAPY Jorge L. Del Aguila Follow this and additional works at: https://digitalcommons.library.tmc.edu/utgsbs_dissertations Part of the Genetics Commons, Genomics Commons, and the Medicine and Health Sciences Commons Recommended Citation Del Aguila, Jorge L., "GENETIC PREDICTORS OF HYPERGLYCEMIA DUE TO HYDROCHLOROTHIAZIDE THERAPY" (2012). The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Dissertations and Theses (Open Access). 233. https://digitalcommons.library.tmc.edu/utgsbs_dissertations/233 This Thesis (MS) is brought to you for free and open access by the The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at DigitalCommons@TMC. It has been accepted for inclusion in The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Dissertations and Theses (Open Access) by an authorized administrator of DigitalCommons@TMC. For more information, please contact [email protected]. GENETIC PREDICTORS OF HYPERGLYCEMIA DUE TO HYDROCHLOROTHIAZIDE THERAPY by Jorge L Del-Aguila APPROVED: ______________________________ Supervisory Professor Eric Boerwinkle, PhD ______________________________ Craig L. Hanis, PhD ______________________________ James E. Hixson, PhD ______________________________ Alanna Morrison, PhD ______________________________ Linda B. Piller, MD, MPH APPROVED: ____________________________ Dean, The University of Texas Graduate School of Biomedical Sciences at Houston GENETIC PREDICTORS OF HYPERGLYCEMIA DUE TO HYDROCHLOROTHIAZIDE THERAPY A THESIS Presented to the Faculty of The University of Texas Health Science Center at Houston and The University of Texas M.
    [Show full text]
  • Network-Based Method for Drug Target Discovery at the Isoform Level
    www.nature.com/scientificreports OPEN Network-based method for drug target discovery at the isoform level Received: 20 November 2018 Jun Ma1,2, Jenny Wang2, Laleh Soltan Ghoraie2, Xin Men3, Linna Liu4 & Penggao Dai 1 Accepted: 6 September 2019 Identifcation of primary targets associated with phenotypes can facilitate exploration of the underlying Published: xx xx xxxx molecular mechanisms of compounds and optimization of the structures of promising drugs. However, the literature reports limited efort to identify the target major isoform of a single known target gene. The majority of genes generate multiple transcripts that are translated into proteins that may carry out distinct and even opposing biological functions through alternative splicing. In addition, isoform expression is dynamic and varies depending on the developmental stage and cell type. To identify target major isoforms, we integrated a breast cancer type-specifc isoform coexpression network with gene perturbation signatures in the MCF7 cell line in the Connectivity Map database using the ‘shortest path’ drug target prioritization method. We used a leukemia cancer network and diferential expression data for drugs in the HL-60 cell line to test the robustness of the detection algorithm for target major isoforms. We further analyzed the properties of target major isoforms for each multi-isoform gene using pharmacogenomic datasets, proteomic data and the principal isoforms defned by the APPRIS and STRING datasets. Then, we tested our predictions for the most promising target major protein isoforms of DNMT1, MGEA5 and P4HB4 based on expression data and topological features in the coexpression network. Interestingly, these isoforms are not annotated as principal isoforms in APPRIS.
    [Show full text]
  • SUPPORTING INFORMATION for Regulation of Gene Expression By
    SUPPORTING INFORMATION for Regulation of gene expression by the BLM helicase correlates with the presence of G4 motifs Giang Huong Nguyen1,2, Weiliang Tang3, Ana I. Robles1, Richard P. Beyer4, Lucas T. Gray5, Judith A. Welsh1, Aaron J. Schetter1, Kensuke Kumamoto1,6, Xin Wei Wang1, Ian D. Hickson2,7, Nancy Maizels5, 3,8 1 Raymond J. Monnat, Jr. and Curtis C. Harris 1Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, U.S.A; 2Department of Medical Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, U.K.; 3Department of Pathology, University of Washington, Seattle, WA U.S.A.; 4 Center for Ecogenetics and Environmental Health, University of Washington, Seattle, WA U.S.A.; 5Department of Immunology and Department of Biochemistry, University of Washington, Seattle, WA U.S.A.; 6Department of Organ Regulatory Surgery, Fukushima Medical University, Fukushima, Japan; 7Cellular and Molecular Medicine, Nordea Center for Healthy Aging, University of Copenhagen, Denmark; 8Department of Genome Sciences, University of WA, Seattle, WA U.S.A. SI Index: Supporting Information for this manuscript includes the following 19 items. A more detailed Materials and Methods section is followed by 18 Tables and Figures in order of their appearance in the manuscript text: 1) SI Materials and Methods 2) Figure S1. Study design and experimental workflow. 3) Figure S2. Immunoblot verification of BLM depletion from human fibroblasts. 4) Figure S3. PCA of mRNA and miRNA expression in BLM-depleted human fibroblasts. 5) Figure S4. qPCR confirmation of mRNA array data. 6) Table S1. BS patient and control detail.
    [Show full text]
  • Hormone and Inhibitor Treatment T47DM Cells Were Used for All Experiments Unless Otherwise Stated
    Extended Data Extended Materials Methods Cell culture; hormone and inhibitor treatment T47DM cells were used for all experiments unless otherwise stated. For hormone induction experiments, cells were grown in RPMI medium without Phenol Red, supplemented with 10% dextran-coated charcoal-treated FBS (DCC/FBS) after 24 h in serum-free conditions; cells were incubated with R5020 (10 nM) or vehicle (ethanol) as described (Vicent et al. 2011). For hormone induction experiments in MCF7 cells a similar procedure was performed; cells were grown in DMEM medium without Phenol Red, supplemented with 10% dextran-coated charcoal-treated FBS (DCC/FBS) after 24 h in serum-free conditions; cells were incubated with Estradiol (10 nM) or vehicle (ethanol). PARG and PARP inhibition were carried out via incubating cells with 5uM TA (tannic acid) or 10uM 3AB (3-amino-benzamide) respectively 1 hour prior to hormone treatment. All transfections were performed using Lipofectamine2000 (Invitrogen) according to manufacturers instructions. PAR-capture ELISA Hormone and or inhibitor treatments were carried out as described, and sample preparation was carried out as follows: At the required time point, cells were washed twice with ice-cold PBS and scraped in lysis buffer (0.4 M NaCl, 1% Triton X-100) plus protease inhibitors. Cell suspensions were then incubated for 30 min on ice with periodic vortexing. The disrupted cell suspension was centrifuged at 10,000g for 10 min at 4°C, and the supernatant was recovered, snap-frozen, and stored at 80°C until required. Ninety-six-well black-walled plates were incubated with 2 ng/mL anti-PAR monoclonal antibody (Trevigen) in 50 mM sodium carbonate (pH 7.6) overnight at 4°C.
    [Show full text]
  • Identifying the Link Between Non-Coding Regulatory Rnas and Phenotypic Severity in a Zebrafish Model of Gmppb Dystroglycanopathy
    The University of Maine DigitalCommons@UMaine Honors College Spring 5-2020 Identifying the Link Between Non-Coding Regulatory RNAs and Phenotypic Severity in a Zebrafish Model of gmppb Dystroglycanopathy Grace Smith Follow this and additional works at: https://digitalcommons.library.umaine.edu/honors Part of the Genetics Commons, Molecular Biology Commons, and the Musculoskeletal Diseases Commons This Honors Thesis is brought to you for free and open access by DigitalCommons@UMaine. It has been accepted for inclusion in Honors College by an authorized administrator of DigitalCommons@UMaine. For more information, please contact [email protected]. IDENTIFYING THE LINK BETWEEN NON-CODING REGULATORY RNAS AND PHENOTYPIC SEVERITY IN A ZEBRAFISH MODEL OF GMPPB DYSTROGLYCANOPATHY by Grace Smith A Thesis Submitted in Partial Fulfillment of the Requirements for a Degree with Honors (Molecular & Cellular Biology) The Honors College University of Maine May 2020 Advisory Committee: Benjamin King, Assistant Professor of Bioinformatics, Advisor Clarissa Henry, Associate Professor of Biological Sciences Melissa Ladenheim, Associate Dean of the Honors College Sally Molloy, Assistant Professor of Genomics and NSFA-Honors Preceptor of Genomics Kristy Townsend, Associate Professor of Neurobiology ABSTRACT Muscular Dystrophy (MD) is characterized by varying severity and time-of-onset by individuals afflicted with the same forms of MD, a phenomenon that is not well understood. MD affects 250,000 individuals in the United States and is characterized by mutations in the dystroglycan complex. gmppb encodes an enzyme that glycosylates dystroglycan, making it functionally active; thus, mutations in gmppb cause dystroglycanopathic MD1. The zebrafish (Danio rerio) is a powerful vertebrate model for musculoskeletal development and disease.
    [Show full text]