Defining and Exploiting the Cytosine Deaminase Activity of Apobec3a on the Extended Epigenome

Total Page:16

File Type:pdf, Size:1020Kb

Defining and Exploiting the Cytosine Deaminase Activity of Apobec3a on the Extended Epigenome University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2018 Defining And Exploiting The Cytosine Deaminase Activity Of Apobec3a On The Extended Epigenome Emily Katharine Schutsky University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Biochemistry Commons, and the Genetics Commons Recommended Citation Schutsky, Emily Katharine, "Defining And Exploiting The Cytosine Deaminase Activity Of Apobec3a On The Extended Epigenome" (2018). Publicly Accessible Penn Dissertations. 2794. https://repository.upenn.edu/edissertations/2794 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/2794 For more information, please contact [email protected]. Defining And Exploiting The Cytosine Deaminase Activity Of Apobec3a On The Extended Epigenome Abstract AID/APOBEC family cytosine deaminases canonically play crucial roles in immunity by converting cytosine to uracil in single-stranded DNA (ssDNA). Outside of this established physiological role, AID/ APOBEC enzymes have also been implicated in the poorly-understood process of DNA demethylation through their proposed deamination of epigenetically-modified cytosine bases like 5-methylcytosine (mC) and 5-hydroxymethylcytosine (hmC). However, there has been no thorough biochemical characterization of AID/APOBEC activity on these substrates, or on the recently-discovered 5-formylcytosine (fC) and 5-carboxylcytosine (caC) to inform this proposed role. Here, we provide the first steady-state kinetic measurements of the most active family member–APOBEC3A(A3A)–against various natural and unnatural modified substrates using a novel, restriction enzyme-based deamination assay. We determined that A3A has poor activity against bulkier oxidized cytosines, such as hmC, fC, and caC, and therefore likely does not contribute substantially to active DNA demethylation via deamination of these bases. By contrast, A3A efficiently deaminates mC in a manner not fully explained by the steric discrimination observed with other AID/APOBEC family members. Because A3A exhibits enzymatic proficiency for C and mC but potently discriminates against x-mCso in vitro, we hypothesized that we could leverage A3A’s biochemical properties to efficiently localize ox-mCs in the genome. We developed APOBEC-Coupled Epigenetic Sequencing (ACE-Seq) – a nondestructive alternative to bisulfite-based methods for base resolution localization of hmC. We first alidatedv this A3A- based method on differentially-modified phage genomes ot demonstrate efficient conversion of C/mC and protection of hmC on a genome scale. We next applied ACE-Seq to mouse embryonic stem cell DNA and found a striking correlation with published hmC data from bisulfite-based methods. eW also sequenced sorted mouse glutamatergic neurons and, using >100-fold less, saw significant correlation with previously published TAB-Seq data from unsorted mouse cortex DNA. Finally, we demonstrate the applicability of nondestructive ACE-Seq to locus-specific analysis with longer (>1 kb) amplicons, which could permit clustering analyses on single reads from larger regions such as enhancers. Altogether, we propose ACE-Seq as a facile alternative to bisulfite-based hmC localization methods, especially for analyzing limiting DNA samples, such as those from small or transient cell populations. Degree Type Dissertation Degree Name Doctor of Philosophy (PhD) Graduate Group Biochemistry & Molecular Biophysics First Advisor Rahul M. Kohli Keywords 5-hydroxymethylcytosine, ACE-Seq, AID/APOBEC, bisulfite-free, deamination, epigenetics Subject Categories Biochemistry | Genetics This dissertation is available at ScholarlyCommons: https://repository.upenn.edu/edissertations/2794 DEFINING AND EXPLOITING THE CYTOSINE DEAMINASE ACTIVITY OF APOBEC3A ON THE EXTENDED EPIGENOME Emily K. Schutsky A DISSERTATION in Biochemistry and Molecular Biophysics Presented to the Faculties of the University of Pennsylvania in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy 2018 Supervisor of Dissertation Signature: ____________________________________ Rahul M. Kohli, M.D., Ph.D., Assistant Professor of Medicine Graduate Group Chairperson Signature: _____________________________________ Kim A. Sharp, Ph.D., Associate Professor of Biochemistry and Biophysics Dissertation Committee Ronen Marmorstein, Ph.D., George W. Raiziss Professor Gregory D. Van Duyne, Ph.D., Jacob Gershon-Cohen Professor Of Medical Science Matthew D. Weitzman, Ph.D., Associate Professor of Pathology and Laboratory Medicine Hongjun Song, Ph.D., Perelman Professor of Neuroscience To my Shepherd, in whom I lack nothing ii ACKNOWLEDGMENT In no way was this work a single-handed effort. Every day, I was physically, emotionally, intellectually, and spiritually supported by so many people who made this culmination possible. All of you believed in me especially in moments when I couldn’t. This is for you. To my labmates, past and present, who individually and uniquely filled each of my days with laughter, resilience, and hope for the next. To my advisor, mentor and friend – Rahul Kohli – who made me feel wholly appreciated, supported, and cared for during each high and low of my graduate career. To my thesis committee – Ronen Marmorstein, Gregory Van Duyne, and Matthew Weitzman – whose advice and suggestions gave me this amazing story to tell. To my collaborators in the Wu lab – Emily Fabyanic, Peng Hu, and Hao Wu – who were a constant source of help and support in making ACE-Seq possible. To the students and faculty in the BMB Graduate Group, and in the BGS graduate program, who created an amazing community of passionate intellectual pursuit. To the city of Philadelphia, who will always hold the dearest place in my heart as a home I didn’t expect, and as a source of brotherly love that has permeated my soul. To even better friends than I could ever have asked for, whose love for me has encouraged me to stand firm in each struggle, big and small. To Ace, the namesake of ACE-Seq and my favorite [canine] nephew, whose love is unconditional as only a dog’s can be. To my family, who has been unceasingly supportive since day one, and who will continue to love and support me no matter where life may lead me. To the God who never fails, and who loves me beyond measure. Thank you. iii ABSTRACT DEFINING AND EXPLOITING THE CYTOSINE DEAMINASE ACTIVITY OF APOBEC3A ON THE EXTENDED EPIGENOME Emily K. Schutsky Rahul M. Kohli, M.D., Ph.D. AID/APOBEC family cytosine deaminases canonically play crucial roles in immunity by converting cytosine to uracil in single-stranded DNA (ssDNA). Outside of this established physiological role, AID/APOBEC enzymes have also been implicated in the poorly-understood process of DNA demethylation through their proposed deamination of epigenetically-modified cytosine bases like 5-methylcytosine (mC) and 5-hydroxymethylcytosine (hmC). However, there has been no thorough biochemical characterization of AID/APOBEC activity on these substrates, or on the recently-discovered 5-formylcytosine (fC) and 5-carboxylcytosine (caC) to inform this proposed role. Here, we provide the first steady-state kinetic measurements of the most active family member–APOBEC3A(A3A)–against various natural and unnatural modified substrates using a novel, restriction enzyme-based deamination assay. We determined that A3A has poor activity against bulkier oxidized cytosines, such as hmC, fC, and caC, and therefore likely does not contribute substantially to active DNA demethylation via deamination of these bases. By contrast, A3A efficiently deaminates mC in a manner not fully explained by the steric discrimination observed with other AID/APOBEC family members. Because A3A exhibits enzymatic proficiency for C and mC but potently discriminates against ox-mCs in vitro, we hypothesized that we could leverage A3A’s biochemical properties to efficiently localize ox-mCs in the genome. We developed APOBEC-Coupled Epigenetic Sequencing (ACE-Seq) – a nondestructive alternative to bisulfite-based methods for base resolution localization of hmC. We first validated this A3A-based method on differentially- iv modified phage genomes to demonstrate efficient conversion of C/mC and protection of hmC on a genome scale. We next applied ACE-Seq to mouse embryonic stem cell DNA and found a striking correlation with published hmC data from bisulfite-based methods. We also sequenced sorted mouse glutamatergic neurons and, using >100-fold less input DNA, saw significant correlation with previously published TAB-Seq data from unsorted mouse cortex DNA. Finally, we demonstrate the applicability of nondestructive ACE-Seq to locus-specific analysis with longer (>1 kb) amplicons, which could permit clustering analyses on single reads from larger regions such as enhancers. Altogether, we propose ACE-Seq as a facile alternative to bisulfite- based hmC localization methods, especially for analyzing limiting DNA samples, such as those from small or transient cell populations. v TABLE OF CONTENTS ACKNOWLEDGMENT .............................................................................................................. iii ABSTRACT ................................................................................................................................... iv LIST OF TABLES ...................................................................................................................... viii LIST OF
Recommended publications
  • Cytoplasmic Activation-Induced Cytidine Deaminase (AID) Exists in Stoichiometric Complex with Translation Elongation Factor 1Α (Eef1a)
    Cytoplasmic activation-induced cytidine deaminase (AID) exists in stoichiometric complex with translation elongation factor 1α (eEF1A) Julien Häsler, Cristina Rada, and Michael S. Neuberger1 Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom Edited by Frederick W. Alt, Howard Hughes Medical Institute, Harvard Medical School, Children’s Hospital Immune Disease Institute, Boston, MA, and approved October 12, 2011 (received for review April 27, 2011) Activation-induced cytidine deaminase (AID) is a B lymphocyte- results reveal that endogenous cytoplasmic AID partakes in a specific DNA deaminase that acts on the Ig loci to trigger antibody complex containing stoichiometric quantities of translation elon- gene diversification. Most AID, however, is retained in the cyto- gation factor 1α (eEF1A), with this association likely implicated in plasm and its nuclear abundance is carefully regulated because the regulation of AID’s intracellular trafficking. off-target action of AID leads to cancer. The nature of the cytosolic AID complex and the mechanisms regulating its release from the Results cytoplasm and import into the nucleus remain unknown. Here, we Flag-Tagging the Endogenous AID Locus in DT40 Cells. We generated show that cytosolic AID in DT40 B cells is part of an 11S complex derivatives of the DT40 B-cell line in which the endogenous AID and, using an endogenously tagged AID protein to avoid overex- locus was modified so as to incorporate a single Flag tag at the pression artifacts, that it is bound in good stoichiometry to the AID N terminus. To allow targeting of both alleles, one targeting translation elongation factor 1 alpha (eEF1A).
    [Show full text]
  • Purification Andsomeproperties of Cytosine Deaminase from Bakers
    Agric. Biol. Chem., 53 (5), 1313-1319, 1989 1313 Purification and SomeProperties of Cytosine Deaminase from Bakers' Yeast Tohoru Katsuragi, Toshihiro Sonoda, Kin'ya Matsumoto, Takuo Sakai and Kenzo Tonomura Laboratory of Fermentation Chemistry, College of Agriculture, University of Osaka Prefecture, Sakai-shi, Osaka 591, Japan Received November 24, 1988 Cytosine deaminase (EC 3.5.4.1) was extracted from commercial compressed bakers' yeast and purified to an almost homogeneous state. The enzyme activity was more than 200U/mg of protein, which was several times higher than reported before. The molecular weight was 41,000 by gel permeation. The pi was at pH4.7. 5-Fluorocytosine, 5-methylcytosine, and creatinine were other substrates for the enzyme.An experiment with inhibitors suggested that the enzyme was an SH- enzyme. The enzyme was unstable to heat, with a half-life of about 0.5hr at 37°C. Characteristics of the enzyme, especially its substrate specificity, were compared with those reported earlier for other cytosine deaminases from bacteria and a mold. Local chemotherapy of cancer with the com- (5MC), a 5-substituted cytosine.4) 5FC, an- bined use of 5-fluorocytosine (5FC) given oral- other 5-substituted cytosine, is deaminated to ly and a cytosine deaminase capsule implant- 5FU in Saccharomyces cerevisiae.5) So, cy- ed locally may be possible.1} However, al- tosine deaminase of bakers' yeast should con- though this approach is successful in animal vert 5FCto 5FU, and could be used in place of experiments,1'2) there are problems when we E. coli cytosine deaminase. Although the yeast use the enzyme from Escherichia coli,3) which enzyme is unstable to heat (at 37.5°C),4) which is thermostable,1'3) and which can deaminate would prevent its use in long-term therapy in 5FC to 5-fluorouracil (5FU).1>3) First, it is the body, it might be stabilized by immobili- difficult to culture the bacteria on a large scale zation or other techniques.
    [Show full text]
  • Molecular Basis of NDT-Mediated Activation of Nucleoside-Based Prodrugs and Application in Suicide Gene Therapy
    biomolecules Article Molecular Basis of NDT-Mediated Activation of Nucleoside-Based Prodrugs and Application in Suicide Gene Therapy Javier Acosta 1,† , Elena Pérez 1,†, Pedro A. Sánchez-Murcia 2, Cristina Fillat 3,4 and Jesús Fernández-Lucas 2,5,* 1 Applied Biotechnology Group, European University of Madrid, c/ Tajo s/n, Villaviciosa de Odón, 28670 Madrid, Spain; [email protected] (J.A.); [email protected] (E.P.) 2 Division of Physiological Chemistry, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/III, A-8010 Graz, Austria; [email protected] 3 Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; cfi[email protected] 4 Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08036 Barcelona, Spain 5 Grupo de Investigación en Ciencias Naturales y Exactas, GICNEX, Universidad de la Costa, CUC, Calle 58 # 55-66 Barranquilla, Colombia * Correspondence: [email protected] † These authors contributed equally to this work. Abstract: Herein we report the first proof for the application of type II 20-deoxyribosyltransferase from Lactobacillus delbrueckii (LdNDT) in suicide gene therapy for cancer treatment. To this end, we first confirm the hydrolytic ability of LdNDT over the nucleoside-based prodrugs 20-deoxy-5- fluorouridine (dFUrd), 20-deoxy-2-fluoroadenosine (dFAdo), and 20-deoxy-6-methylpurine riboside (d6MetPRib). Such activity was significantly increased (up to 30-fold) in the presence of an acceptor nucleobase. To shed light on the strong nucleobase dependence for enzymatic activity, different molecular dynamics simulations were carried out. Finally, as a proof of concept, we tested the LdNDT/dFAdo system in human cervical cancer (HeLa) cells.
    [Show full text]
  • Harnessing the Power of Bacteria in Advancing Cancer Treatment
    International Journal of Molecular Sciences Review Microbes as Medicines: Harnessing the Power of Bacteria in Advancing Cancer Treatment Shruti S. Sawant, Suyash M. Patil, Vivek Gupta and Nitesh K. Kunda * Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Jamaica, NY 11439, USA; [email protected] (S.S.S.); [email protected] (S.M.P.); [email protected] (V.G.) * Correspondence: [email protected]; Tel.: +1-718-990-1632 Received: 20 September 2020; Accepted: 11 October 2020; Published: 14 October 2020 Abstract: Conventional anti-cancer therapy involves the use of chemical chemotherapeutics and radiation and are often non-specific in action. The development of drug resistance and the inability of the drug to penetrate the tumor cells has been a major pitfall in current treatment. This has led to the investigation of alternative anti-tumor therapeutics possessing greater specificity and efficacy. There is a significant interest in exploring the use of microbes as potential anti-cancer medicines. The inherent tropism of the bacteria for hypoxic tumor environment and its ability to be genetically engineered as a vector for gene and drug therapy has led to the development of bacteria as a potential weapon against cancer. In this review, we will introduce bacterial anti-cancer therapy with an emphasis on the various mechanisms involved in tumor targeting and tumor suppression. The bacteriotherapy approaches in conjunction with the conventional cancer therapy can be effective in designing novel cancer therapies. We focus on the current progress achieved in bacterial cancer therapies that show potential in advancing existing cancer treatment options and help attain positive clinical outcomes with minimal systemic side-effects.
    [Show full text]
  • Structural Properties of the Nickel Ions in Urease: Novel Insights Into the Catalytic and Inhibition Mechanisms
    Coordination Chemistry Reviews 190–192 (1999) 331–355 www.elsevier.com/locate/ccr Structural properties of the nickel ions in urease: novel insights into the catalytic and inhibition mechanisms Stefano Ciurli a,*, Stefano Benini b, Wojciech R. Rypniewski b, Keith S. Wilson c, Silvia Miletti a, Stefano Mangani d a Institute of Agricultural Chemistry, Uni6ersity of Bologna, Viale Berti Pichat 10, I-40127 Bologna, Italy b European Molecular Biology Laboratory, c/o DESY, Notkestraße 85, D-22603 Hamburg, Germany c Department of Chemistry, Uni6ersity of York, Heslington, York YO15DD, UK d Department of Chemistry, Uni6ersity of Siena, Pian dei Mantellini 44, I-53100 Siena, Italy Accepted 13 March 1999 Contents Abstract.................................................... 331 1. Biological background ......................................... 332 2. Spectroscopic investigations of the urease active site structure .................. 333 3. Crystallographic studies of the native enzyme ............................ 334 4. Crystallographic studies of urease mutants.............................. 341 5. Crystallographic studies of urease–inhibitor complexes ...................... 345 6. Crystallographic study of a transition state analogue bound to urease.............. 348 7. A novel proposal for the urease mechanism ............................. 350 References .................................................. 353 Abstract This work provides a comprehensive critical summary of urease spectroscopy, crystallogra- phy, inhibitor binding, and site-directed
    [Show full text]
  • Longitudinal Peripheral Blood Transcriptional Analysis of COVID-19 Patients
    medRxiv preprint doi: https://doi.org/10.1101/2020.05.05.20091355; this version posted May 8, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. 1 Longitudinal peripheral blood transcriptional analysis of COVID-19 patients 2 captures disease progression and reveals potential biomarkers 3 Qihong Yan1,5,†, Pingchao Li1,†, Xianmiao Ye1,†, Xiaohan Huang1,5,†, Xiaoneng Mo2, 4 Qian Wang1, Yudi Zhang1, Kun Luo1, Zhaoming Chen1, Jia Luo1, Xuefeng Niu3, Ying 5 Feng3, Tianxing Ji3, Bo Feng3, Jinlin Wang2, Feng Li2, Fuchun Zhang2, Fang Li2, 6 Jianhua Wang1, Liqiang Feng1, Zhilong Chen4,*, Chunliang Lei2,*, Linbing Qu1,*, Ling 7 Chen1,2,3,4,* 8 1Guangzhou Regenerative Medicine and Health-Guangdong Laboratory 9 (GRMH-GDL), Guangdong Laboratory of Computational Biomedicine, Guangzhou 10 Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 11 China 12 2Guangzhou Institute of Infectious Disease, Guangzhou Eighth People’s Hospital, 13 Guangzhou Medical University, Guangzhou, China 14 3State Key Laboratory of Respiratory Disease, National Clinical Research Center for 15 Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated 16 Hospital of Guangzhou Medical University, Guangzhou, China 17 4School of Medicine, Huaqiao University, Xiamen, China 18 5University of Chinese Academy of Science, Beijing, China 19 †These authors contributed equally to this work. 20 *To whom correspondence should be addressed: Ling Chen ([email protected]), 21 Linbing Qu ([email protected]), Chunliang Lei ([email protected]), Zhilong 22 Chen ([email protected]) NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • Functional Genomics Atlas of Synovial Fibroblasts Defining Rheumatoid Arthritis
    medRxiv preprint doi: https://doi.org/10.1101/2020.12.16.20248230; this version posted December 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Functional genomics atlas of synovial fibroblasts defining rheumatoid arthritis heritability Xiangyu Ge1*, Mojca Frank-Bertoncelj2*, Kerstin Klein2, Amanda Mcgovern1, Tadeja Kuret2,3, Miranda Houtman2, Blaž Burja2,3, Raphael Micheroli2, Miriam Marks4, Andrew Filer5,6, Christopher D. Buckley5,6,7, Gisela Orozco1, Oliver Distler2, Andrew P Morris1, Paul Martin1, Stephen Eyre1* & Caroline Ospelt2*,# 1Versus Arthritis Centre for Genetics and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK 2Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland 3Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia 4Schulthess Klinik, Zurich, Switzerland 5Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK 6NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK 7Kennedy Institute of Rheumatology, University of Oxford Roosevelt Drive Headington Oxford UK *These authors contributed equally #corresponding author: [email protected] NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice. 1 medRxiv preprint doi: https://doi.org/10.1101/2020.12.16.20248230; this version posted December 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • A Highly Efficient Non-Viral Process for Programming Mesenchymal Stem
    www.nature.com/scientificreports OPEN A highly efcient non‑viral process for programming mesenchymal stem cells for gene directed enzyme prodrug cancer therapy Yoon Khei Ho*, Jun Yung Woo, Geraldine Xue En Tu, Lih‑Wen Deng & Heng‑Phon Too* Mesenchymal stem cells (MSCs) driven gene‑directed enzyme prodrug therapy has emerged as a potential strategy for cancer treatment. The tumour‑nesting properties of MSCs enable these vehicles to target tumours and metastases with efective therapies. A crucial step in engineering MSCs is the delivery of genetic material with low toxicity and high efciency. Due to the low efciency of current transfection methods, viral vectors are used widely to modify MSCs in preclinical and clinical studies. We show, for the frst time, the high transfection efciency (> 80%) of human adipose tissue derived‑ MSCs (AT‑MSCs) using a cost‑efective and of‑the‑shelf Polyethylenimine, in the presence of histone deacetylase 6 inhibitor and fusogenic lipids. Notably, the phenotypes of MSCs remained unchanged post‑modifcation. AT‑MSCs engineered with a fused transgene, yeast cytosine deaminase::uracil phosphoribosyltransferase (CDy::UPRT) displayed potent cytotoxic efects against breast, glioma, gastric cancer cells in vitro. The efciency of eliminating gastric cell lines were efective even when using 7‑day post‑transfected AT‑MSCs, indicative of the sustained expression and function of the therapeutic gene. In addition, signifcant inhibition of temozolomide resistant glioma tumour growth in vivo was observed with a single dose
    [Show full text]
  • Antibody List
    產品編號 產品名稱 PA569955 1110059E24Rik Polyclonal Antibody PA569956 1110059E24Rik Polyclonal Antibody PA570131 1190002N15Rik Polyclonal Antibody 01-1234-42 123count eBeads Counting Beads MA512242 14.3.3 Pan Monoclonal Antibody (CG15) LFMA0074 14-3-3 beta Monoclonal Antibody (60C10) LFPA0077 14-3-3 beta Polyclonal Antibody PA137002 14-3-3 beta Polyclonal Antibody PA14647 14-3-3 beta Polyclonal Antibody PA515477 14-3-3 beta Polyclonal Antibody PA517425 14-3-3 beta Polyclonal Antibody PA522264 14-3-3 beta Polyclonal Antibody PA529689 14-3-3 beta Polyclonal Antibody MA134561 14-3-3 beta/epsilon/zeta Monoclonal Antibody (3C8) MA125492 14-3-3 beta/zeta Monoclonal Antibody (22-IID8B) MA125665 14-3-3 beta/zeta Monoclonal Antibody (4E2) 702477 14-3-3 delta/zeta Antibody (1H9L19), ABfinity Rabbit Monoclonal 711507 14-3-3 delta/zeta Antibody (1HCLC), ABfinity Rabbit Oligoclonal 702241 14-3-3 epsilon Antibody (5H10L5), ABfinity Rabbit Monoclonal 711273 14-3-3 epsilon Antibody (5HCLC), ABfinity Rabbit Oligoclonal PA517104 14-3-3 epsilon Polyclonal Antibody PA528937 14-3-3 epsilon Polyclonal Antibody PA529773 14-3-3 epsilon Polyclonal Antibody PA575298 14-3-3 eta (Lys81) Polyclonal Antibody MA524792 14-3-3 eta Monoclonal Antibody PA528113 14-3-3 eta Polyclonal Antibody PA529774 14-3-3 eta Polyclonal Antibody PA546811 14-3-3 eta Polyclonal Antibody MA116588 14-3-3 gamma Monoclonal Antibody (HS23) MA116587 14-3-3 gamma Monoclonal Antibody (KC21) PA529690 14-3-3 gamma Polyclonal Antibody PA578233 14-3-3 gamma Polyclonal Antibody 510700 14-3-3 Pan Polyclonal
    [Show full text]
  • Download Download
    Robyn A Lindley. Medical Research Archives vol 8 issue 8. Medical Research Archives REVIEW ARTICLE Review of the mutational role of deaminases and the generation of a cognate molecular model to explain cancer mutation spectra Author Robyn A Lindley1,2 1Department of Clinical Pathology 2GMDx Genomics Ltd, The Victorian Comprehensive Cancer Centre Level 3 162 Collins Street, Faculty of Medicine, Dentistry & Health Sciences Melbourne VIC3000, AUSTRALIA University of Melbourne, Email: [email protected] 305 Gratton Street, Melbourne, VIC 3000, AUSTRALIA Email: [email protected] Correspondence: Robyn A Lindley, Department of Clinical Pathology, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, 305 Gratton Street, Melbourne VIC 3000 AUSTRALIA Mobile: +61 (0) 414209132 Email: [email protected] Abstract Recent developments in somatic mutation analyses have led to the discovery of codon-context targeted somatic mutation (TSM) signatures in cancer genomes: it is now known that deaminase mutation target sites are far more specific than previously thought. As this research provides novel insights into the deaminase origin of most of the somatic point mutations arising in cancer, a clear understanding of the mechanisms and processes involved will be valuable for molecular scientists as well as oncologists and cancer specialists in the clinic. This review will describe the basic research into the mechanism of antigen-driven somatic hypermutation of immunoglobulin variable genes (Ig SHM) that lead to the discovery of TSM signatures, and it will show that an Ig SHM-like signature is ubiquitous in the cancer exome. Most importantly, the data discussed in this review show that Ig SHM-like cancer-associated signatures are highly targeted to cytosine (C) and adenosine (A) nucleotides in a characteristic codon-context fashion.
    [Show full text]
  • WO2019226953A1.Pdf
    ) ( 2 (51) International Patent Classification: Street, Brookline, MA 02446 (US). WILSON, Christo¬ C12N 9/22 (2006.01) pher, Gerard; 696 Main Street, Apartment 311, Waltham, MA 0245 1(US). DOMAN, Jordan, Leigh; 25 Avon Street, (21) International Application Number: Somverville, MA 02143 (US). PCT/US20 19/033 848 (74) Agent: HEBERT, Alan, M. et al. ;Wolf, Greenfield, Sacks, (22) International Filing Date: P.C., 600 Atlanitc Avenue, Boston, MA 02210-2206 (US). 23 May 2019 (23.05.2019) (81) Designated States (unless otherwise indicated, for every (25) Filing Language: English kind of national protection av ailable) . AE, AG, AL, AM, (26) Publication Language: English AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, (30) Priority Data: DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, 62/675,726 23 May 2018 (23.05.2018) US HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KH, KN, KP, 62/677,658 29 May 2018 (29.05.2018) US KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, (71) Applicants: THE BROAD INSTITUTE, INC. [US/US]; MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, 415 Main Street, Cambridge, MA 02142 (US). PRESI¬ OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, DENT AND FELLOWS OF HARVARD COLLEGE SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, [US/US]; 17 Quincy Street, Cambridge, MA 02138 (US).
    [Show full text]