The Effects of ɤ-Glutamyl Hydrolase Modulation on Cancer Treatment and Dna Methylation in Human Cancer Cells

Total Page:16

File Type:pdf, Size:1020Kb

The Effects of ɤ-Glutamyl Hydrolase Modulation on Cancer Treatment and Dna Methylation in Human Cancer Cells THE EFFECTS OF ɤ-GLUTAMYL HYDROLASE MODULATION ON CANCER TREATMENT AND DNA METHYLATION IN HUMAN CANCER CELLS by Sung-Eun Kim A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Nutritional Sciences University of Toronto © Copyright by Sung-Eun Kim 2013 THE EFFECTS OF ɤ-GLUTAMYL HYDROLASE MODULATION ON CANCER TREATMENT AND DNA METHYLATION IN HUMAN CANCER CELLS Sung-Eun Kim Doctor of Philosophy Graduate Department of Nutritional Sciences University of Toronto 2013 Abstract Folate and antifolates are retained in cells by polyglutamylation mediated by folylpolyglutamate synthase (FPGS), and are exported from cells after hydrolysis to monoglutamates by γ-glutamyl hydrolase (GGH). Polyglutamylated (anti)folates are retained in cells longer and are better substrates than their monoglutamate counterparts for intracellular folate-dependent enzymes. GGH modulation may therefore affect chemosensitivity of cancer cells to antifolates and 5- fluorouracil by altering polyglutamylation of antifolates and a specific target intracellular folate cofactor for 5-fluorouracil (5,10-methylenetetrahydrofolate), respectively. We generated an in vitro model of GGH modulation in HCT116 and MDA-MB-435 cells with predictable functional consequences and investigated chemosensitivity to 5-fluorouracil and antifolates. Overall, GGH overexpression decreased chemosensitivity to 5-fluorouracil+ leucovorin and methotrexate, while GGH inhibition increased chemosensitivity to 5-fluorouracil +leucovorin. However, these effects appeared to depend not only on the GGH modulation- induced changes in polyglutamylation of 5,10-methylenetetrahydrofolate and antifolates but also on intracellular folate levels as well as adaptive and compensatory changes in other enzymes ii involved in intracellular folate and antifolate accumulation and metabolism in response to GGH modulation. Polyglutamylation is also important in DNA methylation as polyglutamylated folates are better substrates for methylenetetrahydrofolate reductase and methionine synthase involved in the generation of S-adenosylmethionine, which is a substrate for DNA methylation. We hypothesized that GGH and FPGS modulation may affect DNA methylation at global and gene- specific levels with consequent functional ramifications. GGH and FPGS modulation demonstrated the inverse relationship between GGH activity and global DNA methylation as well as was associated with differential gene expression and altered CpG promoter DNA methylation involved in important biological pathways. Some of the observed altered gene expression appear to be regulated by DNA methylation. Whether or not GGH modulation may be an important determinant of chemosensitivity to 5-fluorouracil- and antifolate-based chemotherapy, and the potential roles of GGH and FPGS modulation in DNA methylation need further exploration. iii Acknowledgments First and foremost, I would like to express my sincere gratitude to my supervisor, Dr. Young-In Kim for the opportunity to complete a Ph.D. degree under his supervision. I truly appreciate his encouragement, guidance and invaluable assistance throughout the project. His knowledge and enthusiasm towards my research have been a true inspiration. I would also like to thank my committee members, Dr. Robert Bruce, Dr. Deborah O’Connor and Dr. Andrew Bognar for their expert knowledge and guidance throughout the course of my research. I would like to extend my gratitude to the collaborators - Dr. Toshinori Hinoue for the bioinformatic analysis of our microarray data, Dr. Daniel Weisenberger and Dr. Peter Laird for advice on the methylation analysis, and Ruth Croxford for the detailed explanations and guidance on the statistics behind the data. I am also grateful to all the past and present lab members for their support and for all the memories we have shared. I am thankful to my friends from the Department of Nutritional Sciences for their support and encouragement throughout my doctorate degree. My deepest thanks are reserved to people dearest and nearest to me. I would like to thank Dr. Mi-Kyung Sung for her continued moral support towards pursuing a graduate degree. I am grateful to all my friends in Toronto and Korea for their attention, encouragement, and support. Special thanks to my family for their love and inspiration throughout my studies. Lastly, to my parents, words cannot describe the gratitude you deserve. I will always appreciate your unwavering love, understanding, and support that have made me who I am today. I would also like to express my gratitude to my grandparents who were always by my side and wished my success. Again, my sincere and lasting gratitude to my father and mother, and it is to them that I dedicate this thesis. iv Table of Contents ACKNOWLEDGMENTS .......................................................................................................... IV TABLE OF CONTENTS .............................................................................................................V LIST OF ABBREVIATIONS .................................................................................................. VII LIST OF TABLES ........................................................................................................................X LIST OF FIGURES ................................................................................................................... XV LIST OF APPENDICES ......................................................................................................... XIX CHAPTER 1: INTRODUCTION .................................................................................................... 1 CHAPTER 2: LITERATURE REVIEW .......................................................................................... 5 2.1 Folate .............................................................................................................................. 6 2.1.1 Chemistry of Folate ................................................................................................. 6 2.1.2 Folate Metabolism and Biochemical Function ....................................................... 7 2.1.3 Intracellular Homeostasis of Folate ..................................................................... 16 2.1.4 Folate and Health ................................................................................................. 27 2.2 Folate and Cancer Risk ................................................................................................ 30 2.3 Folate and Cancer Treatment ....................................................................................... 31 2.3.1 Folate and Chemotherapeutic Agents ................................................................... 31 2.3.2 5-Fluorouracil ....................................................................................................... 34 2.3.3 Methotrexate ......................................................................................................... 37 2.3.4 Pemetrexed ............................................................................................................ 39 2.3.5 Trimetrexate .......................................................................................................... 42 2.3.6 Drug Resistance .................................................................................................... 44 2.4 Epigenetics and Cancer Treatment ............................................................................... 46 2.4.1 Epigenetics ............................................................................................................ 46 2.4.2 DNA Methylation and Cancer .............................................................................. 47 2.4.3 Inhibition of DNA Methylation as Chemotherapeutic Strategy ............................ 52 2.4.4 Epigenetic Silencing and Response to Chemotherapeutic Agents ........................ 53 2.4.5 Analysis of DNA methylation ................................................................................ 54 2.4.6 Folate and Epigenetics ......................................................................................... 56 CHAPTER 3: RATIONALE, HYPOTHESES, AND OBJECTIVES ..................................... 58 CHAPTER 4: STUDY 1 – THE DEVELOPMENT AND VALIDATION OF AN IN VITRO MODEL OF GGH MODULATION AND INVESTIGATION OF THE EFFECT OF GGH v MODULATION ON CHEMOSENSITIVITY OF HUMAN COLON AND BREAST CANCER CELLS TO CHEMOTHERAPEUTICS (PROOF-OF-PRINCIPLE) ...................... 63 4.1 Abstract ......................................................................................................................... 64 4.2 Introduction ................................................................................................................... 65 4.3 Materials and Methods ................................................................................................. 67 4.4 Results... ........................................................................................................................ 73 4.5 Discussion ..................................................................................................................... 87 CHAPTER 5: STUDY 2 – THE EFFECTS OF GGH MODULATION AND FOLATE ON CHEMOSENSITIVITY OF HUMAN COLON AND BREAST CANCER CELLS TO CHEMOTHERAPEUTICS IN IN VITRO AND IN VIVO MODELS ....................................... 93 5.1 Abstract ........................................................................................................................
Recommended publications
  • 1 Evidence for Gliadin Antibodies As Causative Agents in Schizophrenia
    1 Evidence for gliadin antibodies as causative agents in schizophrenia. C.J.Carter PolygenicPathways, 20 Upper Maze Hill, Saint-Leonard’s on Sea, East Sussex, TN37 0LG [email protected] Tel: 0044 (0)1424 422201 I have no fax Abstract Antibodies to gliadin, a component of gluten, have frequently been reported in schizophrenia patients, and in some cases remission has been noted following the instigation of a gluten free diet. Gliadin is a highly immunogenic protein, and B cell epitopes along its entire immunogenic length are homologous to the products of numerous proteins relevant to schizophrenia (p = 0.012 to 3e-25). These include members of the DISC1 interactome, of glutamate, dopamine and neuregulin signalling networks, and of pathways involved in plasticity, dendritic growth or myelination. Antibodies to gliadin are likely to cross react with these key proteins, as has already been observed with synapsin 1 and calreticulin. Gliadin may thus be a causative agent in schizophrenia, under certain genetic and immunological conditions, producing its effects via antibody mediated knockdown of multiple proteins relevant to the disease process. Because of such homology, an autoimmune response may be sustained by the human antigens that resemble gliadin itself, a scenario supported by many reports of immune activation both in the brain and in lymphocytes in schizophrenia. Gluten free diets and removal of such antibodies may be of therapeutic benefit in certain cases of schizophrenia. 2 Introduction A number of studies from China, Norway, and the USA have reported the presence of gliadin antibodies in schizophrenia 1-5. Gliadin is a component of gluten, intolerance to which is implicated in coeliac disease 6.
    [Show full text]
  • Exosomes Confer Chemoresistance to Pancreatic Cancer Cells By
    FULL PAPER British Journal of Cancer (2017) 116, 609–619 | doi: 10.1038/bjc.2017.18 Keywords: chemoresistance; exosomes; pancreatic cancer; ROS; microRNA Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK Girijesh Kumar Patel1, Mohammad Aslam Khan1, Arun Bhardwaj1, Sanjeev K Srivastava1, Haseeb Zubair1, Mary C Patton1, Seema Singh1,2, Moh’d Khushman3 and Ajay P Singh*,1,2 1Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA; 2Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA and 3Department of Interdisciplinary Clinical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA Background: Chemoresistance is a significant clinical problem in pancreatic cancer (PC) and underlying molecular mechanisms still remain to be completely understood. Here we report a novel exosome-mediated mechanism of drug-induced acquired chemoresistance in PC cells. Methods: Differential ultracentrifugation was performed to isolate extracellular vesicles (EVs) based on their size from vehicle- or gemcitabine-treated PC cells. Extracellular vesicles size and subtypes were determined by dynamic light scattering and marker profiling, respectively. Gene expression was examined by qRT-PCR and/or immunoblot analyses, and direct targeting of DCK by miR-155 was confirmed by dual-luciferase 30-UTR reporter assay. Flow cytometry was performed to examine the apoptosis indices and reactive oxygen species (ROS) levels in PC cells using specific dyes. Cell viability was determined using the WST-1 assay. Results: Conditioned media (CM) from gemcitabine-treated PC cells (Gem-CM) provided significant chemoprotection to subsequent gemcitabine toxicity and most of the chemoresistance conferred by Gem-CM resulted from its EVs fraction.
    [Show full text]
  • Sequence Variation in the Dihydrofolate Reductase-Thymidylate Synthase (DHFR-TS) and Trypanothione Reductase (TR) Genes of Trypanosoma Cruzi
    Molecular & Biochemical Parasitology 121 (2002) 33Á/47 www.parasitology-online.com Sequence variation in the dihydrofolate reductase-thymidylate synthase (DHFR-TS) and trypanothione reductase (TR) genes of Trypanosoma cruzi Carlos A. Machado *, Francisco J. Ayala Department of Ecology and Evolutionary Biology, University of California, Irvine, CA 92697-2525, USA Received 15 November 2001; received in revised form 25 January 2002 Abstract Dihydrofolate reductase-thymidylate synthase (DHFR-TS) and trypanothione reductase (TR) are important enzymes for the metabolism of protozoan parasites from the family Trypanosomatidae (e.g. Trypanosoma spp., Leishmania spp.) that are targets of current drug-design studies. Very limited information exists on the levels of genetic polymorphism of these enzymes in natural populations of any trypanosomatid parasite. We present results of a survey of nucleotide variation in the genes coding for those enzymes in a large sample of strains from Trypanosoma cruzi, the agent of Chagas’ disease. We discuss the results from an evolutionary perspective. A sample of 31 strains show 39 silent and five amino acid polymorphisms in DHFR-TS, and 35 silent and 11 amino acid polymorphisms in TR. No amino acid replacements occur in regions that are important for the enzymatic activity of these proteins, but some polymorphisms occur in sites previously assumed to be invariant. The sequences from both genes cluster in four major groups, a result that is not fully consistent with the current classification of T. cruzi in two major groups of strains. Most polymorphisms correspond to fixed differences among the four sequence groups. Two tests of neutrality show that there is no evidence of adaptivedivergence or of selectiveevents having shaped the distribution of polymorphisms and fixed differences in these genes in T.
    [Show full text]
  • DNMT-Focused Library
    DNMT-focused library Medicinal and Computational Chemistry Dept., ChemDiv, Inc., 6605 Nancy Ridge Drive, San Diego, CA 92121 USA, Service: +1 877 ChemDiv, Tel: +1 858-794-4860, Fax: +1 858-794-4931, Email: [email protected] DNA methylation is an essential intracellular event critically involved in a wide range of endogenous processes that play significant role in the foundation of genetic phenomena and diseases. Such epigenetic modifications play a key role in the patho-physiology of many tumors and the current use of agents targeting epigenetic changes has become a topic of intense interest in cancer research. Particularly, DNA methyltransferase (DNMT) is a crucial enzyme for cytosine methylation in DNA [1]. In mammals, DNMTs can be broadly divided into two functional families (DNMT1 and DNMT3). All mammalian DNA methyltransferases are encoded by their own single gene, and consisted of catalytic and regulatory regions (except DNMT2). Via interactions between functional domains in the regulatory or catalytic regions and other adaptors or cofactors, DNA methyltransferases can be localized at selective areas (specific DNA/nucleotide sequence) and linked to specific chromosome status (euchromatin/heterochromatin, various histone modification status). With assistance from UHRF1 and DNMT3L or other factors in DNMT1 and DNMT3a/ DNMT3b, mammalian DNA methyltransferases can be recruited, and then specifically bind to hemimethylated and unmethylated double-stranded DNA sequence to maintain and de novo setup patterns for DNA methylation. Complicated
    [Show full text]
  • Supplemental Materials Supplemental Table 1
    Electronic Supplementary Material (ESI) for RSC Advances. This journal is © The Royal Society of Chemistry 2016 Supplemental Materials Supplemental Table 1. The differentially expressed proteins from rat pancreas identified by proteomics (SAP vs. SO) No. Protein name Gene name ratio P value 1 Metallothionein Mt1m 3.35 6.34E-07 2 Neutrophil antibiotic peptide NP-2 Defa 3.3 8.39E-07 3 Ilf2 protein Ilf2 3.18 1.75E-06 4 Numb isoform o/o rCG 3.12 2.73E-06 5 Lysozyme Lyz2 3.01 5.63E-06 6 Glucagon Gcg 2.89 1.17E-05 7 Serine protease HTRA1 Htra1 2.75 2.97E-05 8 Alpha 2 macroglobulin cardiac isoform (Fragment) 2.75 2.97E-05 9 Myosin IF (Predicted) Myo1f 2.65 5.53E-05 10 Neuroendocrine secretory protein 55 Gnas 2.61 7.60E-05 11 Matrix metallopeptidase 8 Mmp8 2.57 9.47E-05 12 Protein Tnks1bp1 Tnks1bp1 2.53 1.22E-04 13 Alpha-parvin Parva 2.47 1.78E-04 14 C4b-binding protein alpha chain C4bpa 2.42 2.53E-04 15 Protein KTI12 homolog Kti12 2.41 2.74E-04 16 Protein Rab11fip5 Rab11fip5 2.41 2.84E-04 17 Protein Mcpt1l3 Mcpt1l3 2.33 4.43E-04 18 Phospholipase B-like 1 Plbd1 2.33 4.76E-04 Aldehyde dehydrogenase (NAD), cytosolic 19 2.32 4.93E-04 (Fragments) 20 Protein Dpy19l2 Dpy19l2 2.3 5.68E-04 21 Regenerating islet-derived 3 alpha, isoform CRA_a Reg3a 2.27 6.74E-04 22 60S acidic ribosomal protein P1 Rplp1 2.26 7.22E-04 23 Serum albumin Alb 2.25 7.98E-04 24 Ribonuclease 4 Rnase4 2.24 8.25E-04 25 Cct-5 protein (Fragment) Cct5 2.24 8.52E-04 26 Protein S100-A9 S100a9 2.22 9.71E-04 27 Creatine kinase M-type Ckm 2.21 1.00E-03 28 Protein Larp4b Larp4b 2.18 1.25E-03
    [Show full text]
  • Mechanisms for the Inhibition of DNA Methyltransferases by Tea Catechins and Bioflavonoids
    Molecular Pharmacology Fast Forward. Published on July 21, 2005 as DOI: 10.1124/mol.104.008367 Molecular PharmacologyThis article has Fastnot been Forward. copyedited Publishedand formatted. Theon finalJuly version 21, 2005 may differ as doi:10.1124/mol.104.008367from this version. MOL 8367 Mechanisms for the Inhibition of DNA Methyltransferases by Tea Catechins and Bioflavonoids Downloaded from Won Jun Lee, Joong-Youn Shim and Bao Ting Zhu1 Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA (W.J.L., B.T.Z.) molpharm.aspetjournals.org and J. L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707 (J.-Y.S.) at ASPET Journals on September 27, 2021 1 Copyright 2005 by the American Society for Pharmacology and Experimental Therapeutics. Molecular Pharmacology Fast Forward. Published on July 21, 2005 as DOI: 10.1124/mol.104.008367 This article has not been copyedited and formatted. The final version may differ from this version. MOL 8367 Running title: Modulation of DNA methylation by dietary polyphenols Corresponding author: Bao Ting Zhu, Ph.D. Frank and Josie P. Fletcher Professor of Pharmacology and an American Cancer Society Research Scholar, and to whom requests for reprints should be addressed at the Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of South Carolina, Room 617 of Coker Life Sciences Building, 700 Sumter Street, Columbia, SC 29208 (USA). PHONE: 803-777-4802; FAX: 803-777-8356; E-MAIL: [email protected] Number of text pages : 34 Downloaded from Number of tables : 2 Number of figures : 10 Number of references : 39 molpharm.aspetjournals.org Number of words in abstract : 229 Number of words in introduction : 458 Number of words in discussion : 1567 at ASPET Journals on September 27, 2021 2 Molecular Pharmacology Fast Forward.
    [Show full text]
  • Characterization of Timed Changes in Hepatic Copper Concentrations, Methionine Metabolism, Gene Expression, and Global DNA Methylation in the Jackson Toxic Milk Mouse Model Of
    Int. J. Mol. Sci. 2014, 15, 8004-8023; doi:10.3390/ijms15058004 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Article Characterization of Timed Changes in Hepatic Copper Concentrations, Methionine Metabolism, Gene Expression, and Global DNA Methylation in the Jackson Toxic Milk Mouse Model of Wilson Disease Anh Le 1, Noreene M. Shibata 2, Samuel W. French 3, Kyoungmi Kim 4, Kusum K. Kharbanda 5, Mohammad S. Islam 6, Janine M. LaSalle 7, Charles H. Halsted 2, Carl L. Keen 1 and Valentina Medici 2,* 1 Department of Nutrition, University of California Davis, 3135 Meyer Hall, One Shields Avenue, Davis, CA 95616, USA; E-Mails: [email protected] (A.L.); [email protected] (C.L.K.) 2 Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, 4150 V Street, Suite 3500, Sacramento, CA 95817, USA; E-Mails: [email protected] (N.M.S.); [email protected] (C.H.H.) 3 Department of Pathology, UCLA/Harbor Medical Center, 1000 West Carson Street, Torrance, CA 90502, USA; E-Mail: [email protected] 4 Department of Public Health Sciences, Division of Biostatistics, University of California Davis, One Shields Avenue, Med-Sci 1C, Davis, CA 95616, USA; E-Mail: [email protected] 5 Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, VA Medical Center R-151, 4101 Woolworth Avenue, Omaha, NE 68105, USA; E-Mail: [email protected] 6 Department of Medical Microbiology and Immunology, University of California Davis, One Shields Avenue, Tupper Hall, Davis, CA 95616, USA; E-Mail: [email protected] 7 Department of Medical Microbiology and Immunology Genome Center, and MIND Institute, University of California Davis, One Shields Avenue, Tupper Hall, Davis, CA 95616, USA; E-Mail: [email protected] * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +1-916-734-3751; Fax: +1-916-734-7908.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Serine Hydroxymethyl Transferase 1 Stimulates Pro-Oncogenic Cytokine Expression Through Sialic Acid to Promote Ovarian Cancer Tumor Growth and Progression
    OPEN Oncogene (2017) 36, 4014–4024 www.nature.com/onc ORIGINAL ARTICLE Serine hydroxymethyl transferase 1 stimulates pro-oncogenic cytokine expression through sialic acid to promote ovarian cancer tumor growth and progression R Gupta1, Q Yang1, SK Dogra2 and N Wajapeyee1 High-grade serous (HGS) ovarian cancer accounts for 90% of all ovarian cancer-related deaths. However, factors that drive HGS ovarian cancer tumor growth have not been fully elucidated. In particular, comprehensive analysis of the metabolic requirements of ovarian cancer tumor growth has not been performed. By analyzing The Cancer Genome Atlas mRNA expression data for HGS ovarian cancer patient samples, we observed that six enzymes of the folic acid metabolic pathway were overexpressed in HGS ovarian cancer samples compared with normal ovary samples. Systematic knockdown of all six genes using short hairpin RNAs (shRNAs) and follow-up functional studies demonstrated that serine hydroxymethyl transferase 1 (SHMT1) was necessary for ovarian cancer tumor growth and cell migration in culture and tumor formation in mice. SHMT1 promoter analysis identified transcription factor Wilms tumor 1 (WT1) binding sites, and WT1 knockdown resulted in reduced SHMT1 transcription in ovarian cancer cells. Unbiased large-scale metabolomic analysis and transcriptome-wide mRNA expression profiling identified reduced levels of several metabolites of the amino sugar and nucleotide sugar metabolic pathways, including sialic acid N-acetylneuraminic acid (Neu5Ac), and downregulation of pro-oncogenic cytokines interleukin-6 and 8 (IL-6 and IL-8) as unexpected outcomes of SHMT1 loss. Overexpression of either IL-6 or IL-8 partially rescued SHMT1 loss-induced tumor growth inhibition and migration.
    [Show full text]
  • Methotrexate Inhibits the First Committed Step of Purine
    Biochem. J. (1999) 342, 143–152 (Printed in Great Britain) 143 Methotrexate inhibits the first committed step of purine biosynthesis in mitogen-stimulated human T-lymphocytes: a metabolic basis for efficacy in rheumatoid arthritis? Lynette D. FAIRBANKS*, Katarzyna RU$ CKEMANN*1, Ying QIU*2, Catherine M. HAWRYLOWICZ†, David F. RICHARDS†, Ramasamyiyer SWAMINATHAN‡, Bernhard KIRSCHBAUM§ and H. Anne SIMMONDS*3 *Purine Research Laboratory, 5th Floor Thomas Guy House, GKT Guy’s Hospital, London Bridge, London SE1 9RT, U.K., †Department of Respiratory Medicine and Allergy, 5th Floor Thomas Guy House, GKT Guy’s Hospital, London Bridge, London SE1 9RT, U.K., ‡Department of Chemical Pathology, GKT Guy’s Hospital, London Bridge, London SE1 9RT, U.K., and §DG Rheumatic/Autoimmune Diseases, Hoechst Marion Roussel, Deutschland GmbH, D-65926 Frankfurt am Main, Germany The immunosuppressive and anti-inflammatory effects of low- ribosyl-1-pyrophosphate (PP-ribose-P) as the molecular mech- dose methotrexate (MTX) have been related directly to inhibition anism underlying these disparate changes. These results provide of folate-dependent enzymes by polyglutamated derivatives, or the first substantive evidence that the immunosuppressive effects indirectly to adenosine release and\or apoptosis and clonal of low-dose MTX in primary blasting human T-lymphocytes deletion of activated peripheral blood lymphocytes in S-phase. In relate not to the inhibition of the two folate-dependent enzymes this study of phytohaemagglutinin-stimulated primary human T- of purine biosynthesis but to inhibition of the first enzyme, lymphocytes we show that MTX (20 nM to 20 µM) was cytostatic amidophosphoribosyltransferase, thereby elevating PP-ribose-P not cytotoxic, halting proliferation at G".
    [Show full text]
  • DHFR Inhibitors: Reading the Past for Discovering Novel Anticancer Agents
    molecules Review DHFR Inhibitors: Reading the Past for Discovering Novel Anticancer Agents Maria Valeria Raimondi 1,*,† , Ornella Randazzo 1,†, Mery La Franca 1 , Giampaolo Barone 1 , Elisa Vignoni 2, Daniela Rossi 2 and Simona Collina 2,* 1 Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy; [email protected] (O.R.); [email protected] (M.L.F.); [email protected] (G.B.) 2 Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, via Taramelli 12, 27100 Pavia, Italy; [email protected] (E.V.); [email protected] (D.R.) * Correspondence: [email protected] (M.V.R.); [email protected] (S.C.); Tel.: +390-912-389-1915 (M.V.R.); +390-382-987-379 (S.C.) † These Authors contributed equally to this work. Academic Editors: Simona Collina and Mariarosaria Miloso Received: 25 February 2019; Accepted: 20 March 2019; Published: 22 March 2019 Abstract: Dihydrofolate reductase inhibitors are an important class of drugs, as evidenced by their use as antibacterial, antimalarial, antifungal, and anticancer agents. Progress in understanding the biochemical basis of mechanisms responsible for enzyme selectivity and antiproliferative effects has renewed the interest in antifolates for cancer chemotherapy and prompted the medicinal chemistry community to develop novel and selective human DHFR inhibitors, thus leading to a new generation of DHFR inhibitors. This work summarizes the mechanism of action, chemical, and anticancer profile of the DHFR inhibitors discovered in the last six years. New strategies in DHFR drug discovery are also provided, in order to thoroughly delineate the current landscape for medicinal chemists interested in furthering this study in the anticancer field.
    [Show full text]
  • 1611 REGULATION of PYRIMIDINE METABOLISM in PLANTS Chris
    [Frontiers in Bioscience 9, 1611-1625, May 1, 2004] REGULATION OF PYRIMIDINE METABOLISM IN PLANTS 1, 2 1, 3 1, 4 1, 5 1, 6 1, 7 Chris Kafer , Lan Zhou , Djoko Santoso , Adel Guirgis , Brock Weers , Sanggyu Park and Robert Thornburg 1 1 Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, Iowa 50011, 2 BASF Plant Science LLC, 2901 South Loop Drive, Ste 3800, Ames, Iowa 50014, 3 Lan Zhou, Pioneer Hi-Bred International, Inc. 7300 NW 62nd Avenue, PO Box 1004, Johnston, Iowa 50131-1004, 4 Indonesian Biotechnology Research Institute for Estate Crops, Jl, Taman Kencana No 1, Bogor 16151 Indonesia, 5 Institute of Genetic Engineering and Biotechnology, Menofiya University, PO Box 79/22857, Sadat City, Egypt, 6 Department of Biochemistry, University of Iowa, 4/511 Bowen Science Building, Iowa City, Iowa 52242-1109, 7 Division of Life and Environment, College of Natural Resources, Daegu University, Gyongsan City, Gyongbuk, Korea 712-714 TABLE OF CONTENTS 1. Abstract 2. Introduction 3. Pyrimidine metabolic pathways 3.1. De novo pyrimidine biosynthesis 3.1.1. CPSase 3.1.2. ATCase 3.1.3. DHOase 3.1.4. DHODH 3.1.5. UMPS 3.1.6. Intracellular Organization of the de novo Pathway 3.2. Pyrimidine Salvage and Recycling 3.2.1. Cytosine deaminase 3.2.2. Cytidine deaminase 3.2.3. UPRTase 3.3. Pyrimidine Modification 3.3.1. UMP/CMP kinase 3.3.2. NDP kinase 3.3.3. CTP synthase, NDP reductase, dUTPase 3.3.4. Thymidylate synthase/Dihydrofolate reductase 3.4. Pyrimidine Catabolism 4. Regulation of pyrimidine metabolism 4.1.
    [Show full text]