1
|
Rinne J, Niehaus M, Medina-Escobar N, Straube H, Schaarschmidt F, Rugen N, Braun HP, Herde M, Witte CP. Three Arabidopsis UMP kinases have different roles in pyrimidine nucleotide biosynthesis and (deoxy)CMP salvage. THE PLANT CELL 2024; 36:3611-3630. [PMID: 38865437 PMCID: PMC11371195 DOI: 10.1093/plcell/koae170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/09/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Pyrimidine nucleotide monophosphate biosynthesis ends in the cytosol with uridine monophosphate (UMP). UMP phosphorylation to uridine diphosphate (UDP) by UMP KINASEs (UMKs) is required for the generation of all pyrimidine (deoxy)nucleoside triphosphates as building blocks for nucleic acids and central metabolites like UDP-glucose. The Arabidopsis (Arabidopsis thaliana) genome encodes five UMKs and three belong to the AMP KINASE (AMK)-like UMKs, which were characterized to elucidate their contribution to pyrimidine metabolism. Mitochondrial UMK2 and cytosolic UMK3 are evolutionarily conserved, whereas cytosolic UMK1 is specific to the Brassicaceae. In vitro, all UMKs can phosphorylate UMP, cytidine monophosphate (CMP) and deoxycytidine monophosphate (dCMP), but with different efficiencies. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated nuclease 9 (Cas9)-induced null mutants were generated for UMK1 and UMK2, but not for UMK3, since frameshift alleles were lethal for germline cells. However, a mutant with diminished UMK3 activity showing reduced growth was obtained. Metabolome analyses of germinating seeds and adult plants of single- and higher-order mutants revealed that UMK3 plays an indispensable role in the biosynthesis of all pyrimidine (deoxy)nucleotides and UDP-sugars, while UMK2 is important for dCMP recycling that contributes to mitochondrial DNA stability. UMK1 is primarily involved in CMP recycling. We discuss the specific roles of these UMKs referring also to the regulation of pyrimidine nucleoside triphosphate synthesis.
Collapse
Affiliation(s)
- Jannis Rinne
- Department of Molecular Nutrition and Biochemistry of Plants, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Markus Niehaus
- Department of Molecular Nutrition and Biochemistry of Plants, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Nieves Medina-Escobar
- Department of Molecular Nutrition and Biochemistry of Plants, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Henryk Straube
- Department of Molecular Nutrition and Biochemistry of Plants, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Frank Schaarschmidt
- Department of Biostatistics, Institute of Cell Biology and Biophysics, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Nils Rugen
- Department of Plant Proteomics, Institute of Plant Genetics, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Hans-Peter Braun
- Department of Plant Proteomics, Institute of Plant Genetics, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Marco Herde
- Department of Molecular Nutrition and Biochemistry of Plants, Leibniz Universität Hannover, Hannover 30419, Germany
| | - Claus-Peter Witte
- Department of Molecular Nutrition and Biochemistry of Plants, Leibniz Universität Hannover, Hannover 30419, Germany
| |
Collapse
|
2
|
Fehlau M, Kaspar F, Hellendahl KF, Schollmeyer J, Neubauer P, Wagner A. Modular Enzymatic Cascade Synthesis of Nucleotides Using a (d)ATP Regeneration System. Front Bioeng Biotechnol 2020; 8:854. [PMID: 32903716 PMCID: PMC7438870 DOI: 10.3389/fbioe.2020.00854] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
Nucleoside-5'-triphosphates (NTPs) and their analogs are building blocks of DNA and are important compounds in both pharmaceutical and molecular biology applications. Currently, commercially available base or sugar modified NTPs are mainly synthesized chemically. Since the chemical production of NTPs is time-consuming and generally inefficient, alternative approaches are under development. Here we present a simple, efficient and generalizable enzymatic synthesis method for the conversion of nucleosides to NTPs. Our one-pot method is modular, applicable to a wide range of natural and modified nucleotide products and accesses NTPs directly from cheap nucleoside precursors. Nucleoside kinases, nucleoside monophosphate (NMP) kinases and a nucleoside diphosphate (NDP) kinase were applied as biocatalysts. Enzymes with different substrate specificities were combined to produce derivatives of adenosine and cytidine triphosphate with conversions of 4 to 26%. The implementation of a (deoxy)ATP recycling system resulted in a significant increase in the conversion to all NTP products, furnishing 4 different NTPs in quantitative conversion. Natural (deoxy)NTPs were synthesized with 60 to >99% conversion and sugar- and base-modified NTPs were produced with 69 to >99% and 27 to 75% conversion, respectively. The presented method is suitable for the efficient synthesis of a wide range of natural and modified NTPs in a sustainable one-pot process.
Collapse
Affiliation(s)
- Maryke Fehlau
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Berlin, Germany.,BioNukleo GmbH, Berlin, Germany
| | - Felix Kaspar
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Berlin, Germany.,BioNukleo GmbH, Berlin, Germany
| | - Katja F Hellendahl
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Berlin, Germany
| | - Julia Schollmeyer
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Berlin, Germany.,BioNukleo GmbH, Berlin, Germany
| | - Peter Neubauer
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Berlin, Germany
| | - Anke Wagner
- Chair of Bioprocess Engineering, Institute of Biotechnology, Faculty III Process Sciences, Technische Universität Berlin, Berlin, Germany.,BioNukleo GmbH, Berlin, Germany
| |
Collapse
|
3
|
Figueroa DB, Madeen EP, Tillotson J, Richardson P, Cottle L, McCauley M, Landovitz RJ, Andrade A, Hendrix CW, Mayer KH, Wilkin T, Gulick RM, Bumpus NN. Genetic Variation of the Kinases That Phosphorylate Tenofovir and Emtricitabine in Peripheral Blood Mononuclear Cells. AIDS Res Hum Retroviruses 2018; 34:421-429. [PMID: 29455571 DOI: 10.1089/aid.2017.0243] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tenofovir (TFV) disoproxil fumarate and emtricitabine (FTC) are used in combination for HIV treatment and pre-exposure prophylaxis (PrEP). TFV disoproxil fumarate is a prodrug that undergoes diester hydrolysis to TFV. FTC and TFV are nucleoside/nucleotide reverse transcriptase inhibitors that upon phosphorylation to nucleotide triphosphate analogs competitively inhibit HIV reverse transcriptase. We previously demonstrated that adenylate kinase 2, pyruvate kinase, muscle and pyruvate kinase, liver and red blood cell phosphorylate TFV in peripheral blood mononuclear cells (PBMC). To identify the kinases that phosphorylate FTC in PBMC, siRNAs targeted toward kinases that phosphorylate compounds structurally similar to FTC were delivered to PBMC, followed by incubation with FTC and the application of a matrix-assisted laser desorption ionization-mass spectrometry method and ultra high performance liquid chromatography-UV to detect the formation of FTC phosphates. Knockdown of deoxycytidine kinase decreased the formation of FTC-monophosphate, while siRNA targeted toward thymidine kinase 1 decreased the abundance of FTC-diphosphate. Knockdown of either cytidine monophosphate kinase 1 or phosphoglycerate kinase 1 decreased the abundance of FTC-triphosphate. Next-generation sequencing of genomic DNA isolated from 498 HIV-uninfected participants in the HIV Prevention Trials Network 069/AIDS Clinical Trials Group A5305 clinical study, revealed 17 previously unreported genetic variants of TFV or FTC phosphorylating kinases. Of note, four individuals were identified as simultaneous carriers of variants of both TFV and FTC activating kinases. These results identify the specific kinases that activate FTC in PBMC, while also providing further insight into the potential for genetic variation to impact TFV and FTC activation.
Collapse
Affiliation(s)
- Dominique B. Figueroa
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Erin P. Madeen
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph Tillotson
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Paul Richardson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Marybeth McCauley
- FHI 360 Center on AIDS and Community Health, Washington, District of Columbia
| | - Raphael J. Landovitz
- Division of Infectious Diseases, Department of Medicine, UCLA Center for Clinical AIDS Research & Education, Los Angeles, California
| | - Adriana Andrade
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Craig W. Hendrix
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kenneth H. Mayer
- The Fenway Institute, Fenway Health and Harvard Medical School, Boston, Massachusetts
| | - Timothy Wilkin
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Roy M. Gulick
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Namandjé N. Bumpus
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Vinoth A, Thirunalasundari T, Shanmugam M, Uthrakumar A, Suji S, Rajkumar U. Evaluation of DNA methylation and mRNA expression of heat shock proteins in thermal manipulated chicken. Cell Stress Chaperones 2018; 23:235-252. [PMID: 28842808 PMCID: PMC5823805 DOI: 10.1007/s12192-017-0837-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 07/29/2017] [Accepted: 08/02/2017] [Indexed: 01/20/2023] Open
Abstract
Thermal manipulation during embryogenesis has been demonstrated to enhance the thermotolerance capacity of broilers through epigenetic modifications. Heat shock proteins (HSPs) are induced in response to stress for guarding cells against damage. The present study investigates the effect of thermal conditioning during embryogenesis and thermal challenge at 42 days of age on HSP gene and protein expression, DNA methylation and in vitro luciferase assay in brain tissue of Naked Neck (NN) and Punjab Broiler-2 (PB-2) chicken. On the 15th day of incubation, fertile eggs from two breeds, NN and PB-2, were randomly divided in to two groups: control (C)-eggs were incubated under standard incubation conditions, and thermal conditioning (TC)-eggs were exposed to higher incubation temperature (40.5°C) for 3 h on the 15th, 16th, and 17th days of incubation. The chicks obtained from each group were further subdivided and reared under different environmental conditions from the 15th to the 42nd day as normal [N; 25 ± 1 °C, 70% relative humidity (RH)] and heat exposed (HE; 35 ± 1 °C, 50% RH) resulting in four treatment groups (CN, CHE, TCN, and TCHE). The results revealed that HSP promoter activity was stronger in CHE, which had lesser methylation and higher gene expression. The activity of promoter region was lesser in TCHE birds that were thermally manipulated at the embryonic stage, thus reflecting their stress-free condition. This was confirmed by the lower level of mRNA expression of all the HSP genes. In conclusion, thermal conditioning during embryogenesis has a positive impact and improves chicken thermotolerance capacity in postnatal life.
Collapse
Affiliation(s)
- A Vinoth
- Department of Industrial Biotechnology, Bharathidhasan University, Tiruchirappalli, Tamilnadu, 620 024, India
| | - T Thirunalasundari
- Department of Industrial Biotechnology, Bharathidhasan University, Tiruchirappalli, Tamilnadu, 620 024, India
| | - M Shanmugam
- ICAR-Directorate of Poultry Research, Rajendranagar, Hyderabad, Telangana, 500 030, India
| | - A Uthrakumar
- Tamilnadu Veterinary and Animal Sciences University, Chennai, Tamilnadu, India
| | - S Suji
- M.S. Swaminathan Research Institute, Taramani, Chennai, Tamilnadu, India
| | - U Rajkumar
- ICAR-Directorate of Poultry Research, Rajendranagar, Hyderabad, Telangana, 500 030, India.
| |
Collapse
|
5
|
Jordan PC, Stevens SK, Tam Y, Pemberton RP, Chaudhuri S, Stoycheva AD, Dyatkina N, Wang G, Symons JA, Deval J, Beigelman L. Activation Pathway of a Nucleoside Analog Inhibiting Respiratory Syncytial Virus Polymerase. ACS Chem Biol 2017; 12:83-91. [PMID: 28103684 DOI: 10.1021/acschembio.6b00788] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Human respiratory syncytial virus (RSV) is a negative-sense RNA virus and a significant cause of respiratory infection in infants and the elderly. No effective vaccines or antiviral therapies are available for the treatment of RSV. ALS-8176 is a first-in-class nucleoside prodrug inhibitor of RSV replication currently under clinical evaluation. ALS-8112, the parent molecule of ALS-8176, undergoes intracellular phosphorylation, yielding the active 5'-triphosphate metabolite. The host kinases responsible for this conversion are not known. Therefore, elucidation of the ALS-8112 activation pathway is key to further understanding its conversion mechanism, particularly given its potent antiviral effects. Here, we have identified the activation pathway of ALS-8112 and show it is unlike other antiviral cytidine analogs. The first step, driven by deoxycytidine kinase (dCK), is highly efficient, while the second step limits the formation of the active 5'-triphosphate species. ALS-8112 is a 2'- and 4'-modified nucleoside analog, prompting us to investigate dCK recognition of other 2'- and 4'-modified nucleosides. Our biochemical approach along with computational modeling contributes to an enhanced structure-activity profile for dCK. These results highlight an exciting potential to optimize nucleoside analogs based on the second activation step and increased attention toward nucleoside diphosphate and triphosphate prodrugs in drug discovery.
Collapse
Affiliation(s)
- Paul C. Jordan
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Sarah K. Stevens
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Yuen Tam
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Ryan P. Pemberton
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Shuvam Chaudhuri
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Antitsa D. Stoycheva
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Natalia Dyatkina
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Guangyi Wang
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Julian A. Symons
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Jerome Deval
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| | - Leo Beigelman
- Alios BioPharma, Inc., part
of the Janssen Pharmaceutical Companies, South
San Francisco, California, United States
| |
Collapse
|
6
|
Resistance to the nucleotide analogue cidofovir in HPV(+) cells: a multifactorial process involving UMP/CMP kinase 1. Oncotarget 2016; 7:10386-401. [PMID: 26824416 PMCID: PMC4891127 DOI: 10.18632/oncotarget.7006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/05/2016] [Indexed: 12/23/2022] Open
Abstract
Human papillomavirus (HPV) is responsible for cervical cancer, and its role in head and neck carcinoma has been reported. No drug is approved for the treatment of HPV-related diseases but cidofovir (CDV) exhibits selective antiproliferative activity. In this study, we analyzed the effects of CDV-resistance (CDVR) in two HPV(+) (SiHaCDV and HeLaCDV) and one HPV(−) (HaCaTCDV) tumor cell lines. Quantification of CDV metabolites and analysis of the sensitivity profile to chemotherapeutics was performed. Transporters expression related to multidrug-resistance (MRP2, P-gp, BCRP) was also investigated. Alterations of CDV metabolism in SiHaCDV and HeLaCDV, but not in HaCaTCDV, emerged via impairment of UMP/CMPK1 activity. Mutations (P64T and R134M) as well as down-regulation of UMP/CMPK1 expression were observed in SiHaCDV and HeLaCDV, respectively. Altered transporters expression in SiHaCDV and/or HeLaCDV, but not in HaCaTCDV, was also noted. Taken together, these results indicate that CDVR in HPV(+) tumor cells is a multifactorial process.
Collapse
|
7
|
Kasthuri M, El Amri C, Lefort V, Périgaud C, Peyrottes S. Synthesis and study of (R)- and (S)-β-hydroxyphosphonate acyclonucleosides as structural analogues of (S)-HPMPC (cidofovir). NEW J CHEM 2014. [DOI: 10.1039/c4nj00813h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Modification of the acyclic chain highly impacts antiviral activity and recognition by human nmpks.
Collapse
Affiliation(s)
- Mahesh Kasthuri
- IBMM
- UMR 5247 CNRS-UM1-UM2
- Team Nucleosides & Phosphorylated Effectors
- University Montpellier 2
- 34095 Montpellier, France
| | - Chahrazade El Amri
- Sorbonne Universités
- Univ. Paris 06
- UMR 8256 Adaptation biologique et vieillissement
- Enzymologie moléculaire et fonctionnelle
- Equipe Vieillissement Cellulaire Intégré et Inflammation
| | - Valérie Lefort
- Sorbonne Universités
- Univ. Paris 06
- UMR 8256 Adaptation biologique et vieillissement
- Enzymologie moléculaire et fonctionnelle
- Equipe Vieillissement Cellulaire Intégré et Inflammation
| | - Christian Périgaud
- IBMM
- UMR 5247 CNRS-UM1-UM2
- Team Nucleosides & Phosphorylated Effectors
- University Montpellier 2
- 34095 Montpellier, France
| | - Suzanne Peyrottes
- IBMM
- UMR 5247 CNRS-UM1-UM2
- Team Nucleosides & Phosphorylated Effectors
- University Montpellier 2
- 34095 Montpellier, France
| |
Collapse
|
8
|
Sjuvarsson E, Damaraju VL, Mowles D, Sawyer MB, Tiwari R, Agarwal HK, Khalil A, Hasabelnaby S, Goudah A, Nakkula RJ, Barth RF, Cass CE, Eriksson S, Tjarks W. Cellular influx, efflux, and anabolism of 3-carboranyl thymidine analogs: potential boron delivery agents for neutron capture therapy. J Pharmacol Exp Ther 2013; 347:388-97. [PMID: 24006340 DOI: 10.1124/jpet.113.207464] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
3-[5-{2-(2,3-Dihydroxyprop-1-yl)-o-carboran-1-yl}pentan-1-yl]thymidine (N5-2OH) is a first generation 3-carboranyl thymidine analog (3CTA) that has been intensively studied as a boron-10 ((10)B) delivery agent for neutron capture therapy (NCT). N5-2OH is an excellent substrate of thymidine kinase 1 and its favorable biodistribution profile in rodents led to successful preclinical NCT of rats bearing intracerebral RG2 glioma. The present study explored cellular influx and efflux mechanisms of N5-2OH, as well as its intracellular anabolism beyond the monophosphate level. N5-2OH entered cultured human CCRF-CEM cells via passive diffusion, whereas the multidrug resistance-associated protein 4 appeared to be a major mediator of N5-2OH monophosphate efflux. N5-2OH was effectively monophosphorylated in cultured murine L929 [thymidine kinase 1 (TK1(+))] cells whereas formation of N5-2OH monophosphate was markedly lower in L929 (TK1(-)) cell variants. Further metabolism to the di- and triphosphate forms was not observed in any of the cell lines. Regardless of monophosphorylation, parental N5-2OH was the major intracellular component in both TK1(+) and TK1(-) cells. Phosphate transfer experiments with enzyme preparations showed that N5-2OH monophosphate, as well as the monophosphate of a second 3-carboranyl thymidine analog [3-[5-(o-carboran-1-yl)pentan-1-yl]thymidine (N5)], were not substrates of thymidine monophosphate kinase. Surprisingly, N5-diphosphate was phosphorylated by nucleoside diphosphate kinase although N5-triphosphate apparently was not a substrate of DNA polymerase. Our results provide valuable information on the cellular metabolism and pharmacokinetic profile of 3-carboranyl thymidine analogs.
Collapse
Affiliation(s)
- Elena Sjuvarsson
- Department of Anatomy, Physiology, and Biochemistry, The Swedish University of Agricultural Sciences, Biomedical Center, Uppsala, Sweden (E.S., S.E.); Department of Oncology, University of Alberta, Edmonton, Alberta, Canada (V.L.D., D.M., M.B.S., C.E.C); Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio (R.T., H.K.A., A.K., S.H., A.G., W.T.); Chemistry Department, Faculty of Science, Zagazig University, Zagazig, Egypt (A.K.); Division of Pharmaceutical Organic Chemistry, College of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt (S.H.); Division of Pharmacology, College of Veterinary Medicine, Cairo University, Giza, Egypt (A.G.); and Department of Pathology, The Ohio State University, Columbus, Ohio (R.J.N., R.F.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Murphy JM, Armijo AL, Nomme J, Lee CH, Smith QA, Li Z, Campbell DO, Liao HI, Nathanson DA, Austin WR, Lee JT, Darvish R, Wei L, Wang J, Su Y, Damoiseaux R, Sadeghi S, Phelps ME, Herschman HR, Czernin J, Alexandrova AN, Jung ME, Lavie A, Radu CG. Development of new deoxycytidine kinase inhibitors and noninvasive in vivo evaluation using positron emission tomography. J Med Chem 2013; 56:6696-708. [PMID: 23947754 DOI: 10.1021/jm400457y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Combined inhibition of ribonucleotide reductase and deoxycytidine kinase (dCK) in multiple cancer cell lines depletes deoxycytidine triphosphate pools leading to DNA replication stress, cell cycle arrest, and apoptosis. Evidence implicating dCK in cancer cell proliferation and survival stimulated our interest in developing small molecule dCK inhibitors. Following a high throughput screen of a diverse chemical library, a structure-activity relationship study was carried out. Positron Emission Tomography (PET) using (18)F-L-1-(2'-deoxy-2'-FluoroArabinofuranosyl) Cytosine ((18)F-L-FAC), a dCK-specific substrate, was used to rapidly rank lead compounds based on their ability to inhibit dCK activity in vivo. Evaluation of a subset of the most potent compounds in cell culture (IC50 = ∼1-12 nM) using the (18)F-L-FAC PET pharmacodynamic assay identified compounds demonstrating superior in vivo efficacy.
Collapse
Affiliation(s)
- Jennifer M Murphy
- Department of Molecular and Medical Pharmacology, §Ahmanson Translational Imaging Division, ⊥Department of Chemistry and Biochemistry, #California NanoSystems Institute, △Department of Biological Chemistry, University of California, Los Angeles , 650 Charles E. Young Dr. S., Los Angeles, California 90095, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Bardhan K, Liu K. Epigenetics and colorectal cancer pathogenesis. Cancers (Basel) 2013; 5:676-713. [PMID: 24216997 PMCID: PMC3730326 DOI: 10.3390/cancers5020676] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) develops through a multistage process that results from the progressive accumulation of genetic mutations, and frequently as a result of mutations in the Wnt signaling pathway. However, it has become evident over the past two decades that epigenetic alterations of the chromatin, particularly the chromatin components in the promoter regions of tumor suppressors and oncogenes, play key roles in CRC pathogenesis. Epigenetic regulation is organized at multiple levels, involving primarily DNA methylation and selective histone modifications in cancer cells. Assessment of the CRC epigenome has revealed that virtually all CRCs have aberrantly methylated genes and that the average CRC methylome has thousands of abnormally methylated genes. Although relatively less is known about the patterns of specific histone modifications in CRC, selective histone modifications and resultant chromatin conformation have been shown to act, in concert with DNA methylation, to regulate gene expression to mediate CRC pathogenesis. Moreover, it is now clear that not only DNA methylation but also histone modifications are reversible processes. The increased understanding of epigenetic regulation of gene expression in the context of CRC pathogenesis has led to development of epigenetic biomarkers for CRC diagnosis and epigenetic drugs for CRC therapy.
Collapse
Affiliation(s)
- Kankana Bardhan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, and Cancer Center, Georgia Regents University, Augusta, GA 30912, USA.
| | | |
Collapse
|
11
|
El Amri C, Martin AR, Vasseur JJ, Smietana M. Borononucleotides as substrates/binders for human NMP kinases: enzymatic and spectroscopic evaluation. Chembiochem 2012; 13:1605-12. [PMID: 22733592 DOI: 10.1002/cbic.201200199] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Indexed: 11/06/2022]
Abstract
Borononucleotides are a family of natural nucleotide monophosphate analogues with a 5'-boronic acid function. As B-O-P linkages are known to be unstable in solution, we evaluated the ability of borononucleotides to be recognized by nucleoside monophosphate kinases and eventually foil the phosphorylation process. In this context, and with the idea of probing the influence of their size, shape, and flexibility, a library of borononucleotides were synthetized starting from the borononucleotide analogue of thymidine, which was shown to behave as a slow substrate of human TMP kinase. This study thus constitutes a good starting point for the development of new monophosphate mimics as potential substrates or ligands for NMP kinases.
Collapse
Affiliation(s)
- Chahrazade El Amri
- Groupe d'Enzymologie Moléculaire et Fonctionnelle, UR4-UPMC, Université Pierre et Marie Curie, Sorbonne Universités, case courrier 256, 7, quai St Bernard, 75252 Paris Cedex 05, France.
| | | | | | | |
Collapse
|
12
|
Evaluation of a UCMK/dCK fusion enzyme for gemcitabine-mediated cytotoxicity. Biochem Biophys Res Commun 2011; 416:199-204. [PMID: 22093835 DOI: 10.1016/j.bbrc.2011.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/04/2011] [Indexed: 11/22/2022]
Abstract
While gemcitabine (2'-2'-difluoro-2'-deoxycytidine, dFdC) displays wide-ranging antineoplastic activity as a single agent, variable response rates and poor intracellular metabolism often limit its clinical efficacy. In an effort to enhance dFdC cytotoxicity and help normalize response rates, we created a bifunctional fusion enzyme that combines the enzymatic activities of deoxycytidine kinase (dCK) and uridine/cytidine monophosphate kinase (UCMK) in a single polypeptide. Our goal was to evaluate whether the created fusion could induce beneficial, functional changes toward dFdC, expedite dFdC conversion to its active antimetabolites and consequently amplify cell dFdC sensitivity. While kinetic analyses revealed the UCMK/dCK fusion enzyme to possess both native activities, the fusion rendered cells sensitive to the cytotoxic effects of dFdC at the same level as dCK expression alone. These results suggest that increased wild-type UCMK expression does not provide a significant enhancement in dFdC-mediated cytotoxicity and may warrant the implementation of studies aimed at engineering UCMK variants with improved activity toward gemcitabine monophosphate.
Collapse
|
13
|
Epigenetics and chemoresistance in colorectal cancer: an opportunity for treatment tailoring and novel therapeutic strategies. Drug Resist Updat 2011; 14:280-96. [PMID: 21955833 DOI: 10.1016/j.drup.2011.08.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 08/09/2011] [Accepted: 08/13/2011] [Indexed: 02/07/2023]
Abstract
Colorectal cancer is the second leading cause of cancer-related deaths in the world. Despite many therapeutic opportunities, prognosis remains dismal for patients with metastatic disease, and a significant portion of early-stage patients develop recurrence after chemotherapy. Epigenetic gene regulation is a major mechanism of cancer initiation and progression, through the inactivation of several tumor suppressor genes. Emerging evidence indicates that epigenetics may also play a key role in the development of chemoresistance. In the present review, we summarize epigenetic mechanisms triggering resistance to three commonly used agents in colorectal cancer: 5-fluorouracil, irinotecan and oxaliplatin. Those epigenetic biomarkers may help stratify colorectal cancer patients and develop a tailored therapeutic approach. In addition, epigenetic modifications are reversible through specific drugs: histone-deacetylase and DNA-methyl-transferase inhibitors. Preclinical studies suggest that these drugs may reverse chemoresistance in colorectal tumors. In conclusion, an epigenetic approach to colorectal cancer chemoresistance may pave the way to personalized treatment and to innovative therapeutic strategies.
Collapse
|
14
|
Gallier F, Alexandre JAC, El Amri C, Deville-Bonne D, Peyrottes S, Périgaud C. 5′,6′-Nucleoside Phosphonate Analogues Architecture: Synthesis and Comparative Evaluation towards Metabolic Enzymes. ChemMedChem 2011; 6:1094-106. [DOI: 10.1002/cmdc.201100068] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 04/04/2011] [Indexed: 01/27/2023]
|
15
|
Hu R, Lam W, Hsu CH, Cheng YC. UMP/CMPK is not the critical enzyme in the metabolism of pyrimidine ribonucleotide and activation of deoxycytidine analogs in human RKO cells. PLoS One 2011; 6:e19490. [PMID: 21559290 PMCID: PMC3086915 DOI: 10.1371/journal.pone.0019490] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 04/07/2011] [Indexed: 11/26/2022] Open
Abstract
Background Human UMP/CMP kinase was identified based on its enzymatic activity in vitro. The role of this protein is considered critical for the maintenance of pyrimidine nucleotide pool profile and for the metabolism of pyrimidine analogs in cells, based on the in vitro study of partially purified enzyme and recombinant protein. However, no detailed study has yet addressed the role of this protein in nucleotide metabolism in cells. Methodology/Principal Findings Two stable cell lines in which UMP/CMP kinase (mRNA: AF087865, EC 2.7.4.14) can be either up-regulated or down-regulated were developed using Tet-On Gene Expression Systems. The amount and enzymatic activity of UMP/CMP kinase extracted from these two cell lines can be induced up by 500% or down by 95–98%. The ribonucleotides of endogenous pyrimidine as well as the metabolism of exogenous natural pyrimidine nucleosides and their analogs were not susceptible to the altered amount of UMP/CMP kinase in these two stable RKO cell lines. The level of incorporation of pyrimidine nucleoside analogs, such as gemcitabine (dFdC) and troxacitabine (L-OddC), into cellular DNA and their potency in inhibiting cell growth were not significantly altered by up-regulation or down-regulation of UMP/CMP kinase expression in cells. Conclusions/Significance The UMP/CMP kinase (EC 2.7.4.14) expressed in RKO cells is not critical for the phosphorylation of (d)CMP and the maintenance of natural nucleotide pools. It also does not play an important role in the activation of dFdC and L-OddC. The increase by 500% or decrease by 95–98% in the levels of UMP/CMP kinase do not affect steady state levels of dFdC and L-OddC in RKO cells. Overall, the activity and possible mechanisms of recombinant UMP/CMP kinase expressed in the in vitro system can not be extended to that of UMP/CMP kinase expressed in a cell system or an in vivo system.
Collapse
Affiliation(s)
- Rong Hu
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Wing Lam
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Chih-Hung Hsu
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
16
|
Foster MS, Oldham CD, May SW. Looking glass mechanism-based inhibition of peptidylglycine α-amidating monooxygenase. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/j.tetasy.2011.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Topalis D, Pradère U, Roy V, Caillat C, Azzouzi A, Broggi J, Snoeck R, Andrei G, Lin J, Eriksson S, Alexandre JAC, El-Amri C, Deville-Bonne D, Meyer P, Balzarini J, Agrofoglio LA. Novel Antiviral C5-Substituted Pyrimidine Acyclic Nucleoside Phosphonates Selected as Human Thymidylate Kinase Substrates. J Med Chem 2010; 54:222-32. [DOI: 10.1021/jm1011462] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Dimitri Topalis
- Groupe d’Enzymologie Moléculaire et Fonctionnelle, UR4-UPMC, Université Pierre et Marie Curie, Sorbonne Universités, case courrier 256, 7, quai St Bernard, 75252 Paris Cedex 05, France
| | - Ugo Pradère
- Institut de Chimie Organique et Analytique, Centre National de Recherche Scientifique Unité Mixte de Recherche 6005, Université d’Orléans, 45067 Orléans, France
| | - Vincent Roy
- Institut de Chimie Organique et Analytique, Centre National de Recherche Scientifique Unité Mixte de Recherche 6005, Université d’Orléans, 45067 Orléans, France
| | - Christophe Caillat
- Laboratoire d’Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique UPR 3082, 91198 Gif-sur-Yvette Cedex, France
| | - Ahmed Azzouzi
- Institut de Chimie Organique et Analytique, Centre National de Recherche Scientifique Unité Mixte de Recherche 6005, Université d’Orléans, 45067 Orléans, France
| | - Julie Broggi
- Institut de Chimie Organique et Analytique, Centre National de Recherche Scientifique Unité Mixte de Recherche 6005, Université d’Orléans, 45067 Orléans, France
| | - Robert Snoeck
- REGA Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Graciela Andrei
- REGA Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jay Lin
- Department of Veterinary Medical Chemistry, Swedish University of Agricultural Sciences, Box 575, Biomedical Center, S-751 24 Uppsala, Sweden
| | - Staffan Eriksson
- Department of Veterinary Medical Chemistry, Swedish University of Agricultural Sciences, Box 575, Biomedical Center, S-751 24 Uppsala, Sweden
| | - Julie A. C. Alexandre
- Groupe d’Enzymologie Moléculaire et Fonctionnelle, UR4-UPMC, Université Pierre et Marie Curie, Sorbonne Universités, case courrier 256, 7, quai St Bernard, 75252 Paris Cedex 05, France
| | - Chahrazade El-Amri
- Groupe d’Enzymologie Moléculaire et Fonctionnelle, UR4-UPMC, Université Pierre et Marie Curie, Sorbonne Universités, case courrier 256, 7, quai St Bernard, 75252 Paris Cedex 05, France
| | - Dominique Deville-Bonne
- Groupe d’Enzymologie Moléculaire et Fonctionnelle, UR4-UPMC, Université Pierre et Marie Curie, Sorbonne Universités, case courrier 256, 7, quai St Bernard, 75252 Paris Cedex 05, France
| | - Philippe Meyer
- Laboratoire d’Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique UPR 3082, 91198 Gif-sur-Yvette Cedex, France
| | - Jan Balzarini
- REGA Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Luigi A. Agrofoglio
- Institut de Chimie Organique et Analytique, Centre National de Recherche Scientifique Unité Mixte de Recherche 6005, Université d’Orléans, 45067 Orléans, France
| |
Collapse
|
18
|
Clozapine treatment causes oxidation of proteins involved in energy metabolism in lymphoblastoid cells: a possible mechanism for antipsychotic-induced metabolic alterations. J Psychiatr Pract 2010; 16:325-33. [PMID: 20859109 DOI: 10.1097/01.pra.0000388627.36781.6a] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
There is increasing concern about the serious metabolic side effects and neurotoxicity caused by atypical (second-generation) antipsychotics. In a previous study by our group (Walss-Bass et al. Int J Neuropsychopharmacol 2008;11:1097-104), using a novel proteomic approach, we showed that clozapine treatment in SKNSH cells induces oxidation of proteins involved in energy metabolism, leading us to hypothesize that protein oxidation could be a mechanism by which atypical antipsychotics increase the risk for metabolic alterations. In this study, the same proteomic approach was used to identify specific proteins oxidized after clozapine treatment in lymphoblastoid cell lines from patients with schizophrenia and normal controls. Cells were treated with 0 and 20 μM clozapine for 24 hours and protein extracts were labeled with 6-iodoacetamide fluorescein (6-IAF). The lack of incorporation of 6-IAF into the thiol group of cysteine residues is an indicator of protein oxidation. Labeled proteins were exposed to two dimensional electrophoresis, and differential protein labeling was assessed. Increased oxidation after clozapine treatment was observed in 9 protein spots (P<0.05). The following 7 proteins were identified by high-performance liquid chromatography-electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS) in those 9 spots: enolase, triosephosphate isomerase (TPI), glyceraldehyde-3-phosphate dehydrogenase (GAPD), Rho GDP dissociation inhibitor (GDI), cofilin, uridine monophosphate/ cytidine monophosphate (UMP-CMP) kinase, and translation elongation factor. Several of these proteins play important roles in energy metabolism and mitochondrial function. These results further support the hypothesis that oxidative stress may be a mechanism by which antipsychotics increase the risk of metabolic syndrome and diabetes.
Collapse
|
19
|
Deville-Bonne D, El Amri C, Meyer P, Chen Y, Agrofoglio LA, Janin J. Human and viral nucleoside/nucleotide kinases involved in antiviral drug activation: structural and catalytic properties. Antiviral Res 2010; 86:101-20. [PMID: 20417378 DOI: 10.1016/j.antiviral.2010.02.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 01/31/2010] [Accepted: 02/01/2010] [Indexed: 12/11/2022]
Abstract
Antiviral nucleoside and nucleotide analogs, essential for the treatment of viral infections in the absence of efficient vaccines, are prodrug forms of the active compounds that target the viral DNA polymerase or reverse transcriptase. The activation process requires several successive phosphorylation steps catalyzed by different kinases, which are present in the host cell or encoded by some of the viruses. These activation reactions often are rate-limiting steps and are thus open to improvement. We review here the structural and enzymatic properties of the enzymes that carry out the activation of analogs used in therapy against human immunodeficiency virus and against DNA viruses such as hepatitis B, herpes and poxviruses. Four major classes of drugs are considered: thymidine analogs, non-natural L-nucleosides, acyclic nucleoside analogs and acyclic nucleoside phosphonate analogs. Their efficiency as drugs depends both on the low specificity of the viral polymerase that allows their incorporation into DNA, but also on the ability of human/viral kinases to provide the activated triphosphate active forms at a high concentration at the right place. Two distinct modes of action are considered, depending on the origin of the kinase (human or viral). If the human kinases are house-keeping enzymes that belong to the metabolic salvage pathway, herpes and poxviruses encode for related enzymes. The structures, substrate specificities and catalytic properties of each of these kinases are discussed in relation to drug activation.
Collapse
Affiliation(s)
- Dominique Deville-Bonne
- Enzymologie Moléculaire et Fonctionnelle, UR4 Université Pierre et Marie Curie, 7 quai St Bernard, 75252 Paris Cedex 05, France.
| | | | | | | | | | | |
Collapse
|
20
|
Identification and biochemical characterization of a unique Mn2+-dependent UMP kinase from Helicobacter pylori. Arch Microbiol 2010; 192:739-46. [PMID: 20602229 DOI: 10.1007/s00203-010-0600-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 05/20/2010] [Accepted: 06/10/2010] [Indexed: 10/19/2022]
Abstract
Uridine monophosphate (UMP) kinase converts UMP to the corresponding UDP in the presence of metal ions and ATP and is allosterically regulated by nucleotides such as UTP and GTP. Although the UMP kinase reported to date is Mg(2+)-dependent, we found in this study that the UMP kinase of Helicobacter pylori had a preference for Mn(2+) over Mg(2+), which may be related to a conformational difference between the Mn(2+)-bound and Mg(2+)-bound UMP kinase. Similar to previous findings, the UMP kinase activity of H. pylori UMP kinase was inhibited by UTP and activated by GTP. However, a relatively low GTP concentration (0.125 mM) was required to activate H. pylori UMP kinase to a level similar to other bacterial UMP kinases using a higher GTP concentration (0.5 mM). In addition, depending on the presence of either Mg(2+) or Mn(2+), a significant difference in the level of GTP activation was observed. It is therefore hypothesized that the Mg(2+)-bound and Mn(2+)-bound H. pylori UMP kinase may be activated by GTP through different mechanisms.
Collapse
|
21
|
Liou JY, Lai HR, Hsu CH, Chang WL, Hsieh MJ, Huang YC, Cheng YC. Modulation of human UMP/CMP kinase affects activation and cellular sensitivity of deoxycytidine analogs. Biochem Pharmacol 2009; 79:381-8. [PMID: 19765547 DOI: 10.1016/j.bcp.2009.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 10/20/2022]
Abstract
Deoxycytidine analogs are an important class of clinically active antiviral and anticancer agents. The stepwise phosphorylation of these analogs to triphosphate metabolites is crucial for biological action. Human UMP/CMP kinase (UMP/CMPK; cytidylate kinase; EC 2.7.4.14) is thought to be responsible for phosphorylation of UMP, CMP, and dCMP and may also play an important role in the activation of pyrimidine analogs. However, no evidence has verified this notion in intact cells. In this study we explored the functional roles of UMP/CMPK in natural pyrimidine synthesis and metabolism of deoxycytidine analogs, as well as 5-FU in HeLa S3 and HCT8 cells. The amounts of UMP/CMPK protein in different cell lines correlated with UMP, CMP, and dCMP kinase activities and amounts of UMP/CMPK RNA. Modulation of UMP/CMPK by overexpression or down-regulation had no impact on natural pyrimidine nucleotides and cell growth. However, down-regulating UMP/CMPK expression by siRNA led to a decrease in the formation of the triphosphate metabolites, resulting in cellular resistance to these analogs. More diphosphate and triphosphate metabolites of deoxycytidine analogs were detected and cellular sensitivity to these agents was increased in the UMP/CMPK-overexpressing cells. This study indicates that the second step enzyme (UMP/CMPK) is responsible for the phosphorylation of pyrimidine analogs and also has an impact on cellular sensitivity to these analogs in those cell lines.
Collapse
Affiliation(s)
- Jieh-Yuan Liou
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC.
| | | | | | | | | | | | | |
Collapse
|
22
|
Humeniuk R, Menon LG, Mishra PJ, Gorlick R, Sowers R, Rode W, Pizzorno G, Cheng YC, Kemeny N, Bertino JR, Banerjee D. Decreased levels of UMP kinase as a mechanism of fluoropyrimidine resistance. Mol Cancer Ther 2009; 8:1037-44. [PMID: 19383847 DOI: 10.1158/1535-7163.mct-08-0716] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
5-Fluorouracil (5-FU) continues to be widely used for treatment of gastrointestinal cancers. Because many tumors show primary or acquired resistance, it is important to understand the molecular basis underlying the mechanism of resistance to 5-FU. In addition to its effect on thymidylate synthase inhibition and DNA synthesis, 5-FU may also influence RNA metabolism. Our previous studies revealed that colorectal cancer cells resistant to bolus 5-FU (HCT-8/4hFU) showed significantly decreased incorporation of the drug into RNA. Resistance to bolus 5-FU was associated with lower expression of UMP kinase (UMPK), an enzyme that plays an important role in the activation of 5-FU to 5-FUTP and its incorporation into RNA. Activities of other 5-FU-metabolizing enzymes (e.g., thymidine kinase, uridine phosphorylase, thymidine phosphorylase, and orotate phosphoribosyltransferase) remained unchanged between sensitive and resistant cell lines. Herein, we show that UMPK down-regulation in 5-FU-sensitive cells (HCT-8/P) induces resistance to bolus 5-FU treatment. Moreover, HCT-8/4hFU cells are even more cross-resistant to treatment with 5-fluorouridine, consistent with the current understanding of 5-fluorouridine as a RNA-directed drug. Importantly, colorectal cancer hepatic metastases isolated from patients clinically resistant to weekly bolus 5-FU/leucovorin treatment exhibited decreased mRNA expression of UMPK but not thymidylate synthase or dihydropyrimidine dehydrogenase compared with tumor samples of patients not previously exposed to 5-FU. Our findings provide new insights into the mechanisms of acquired resistance to 5-FU in colorectal cancer and implicate UMPK as an important mechanism of clinical resistance to pulse 5-FU treatment in some patients.
Collapse
Affiliation(s)
- Rita Humeniuk
- The Graduate School of Biomedical Sciences, Department of Pharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 08903, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Topalis D, Alvarez K, Barral K, Munier-Lehmann H, Schneider B, Véron M, Guerreiro C, Mulard L, El-Amri C, Canard B, Deville-Bonne D. Acyclic phosphonate nucleotides and human adenylate kinases: impact of a borano group on alpha-P position. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2008; 27:319-31. [PMID: 18404568 DOI: 10.1080/15257770801941952] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Adenylate kinases are involved in the activation of antiviral drugs such as the acyclic phosphonates analogs PMEA and (R)PMPA. We examine the in vitro phosphorylation of PMEA and PMPA bearing a borano- or a H- group on the phosphorus atom. The alpha-borano or alpha-H on PMEA and PMPA were detrimental to the activity of recombinant human AMP kinases 1 and 2. Docking PMEA to the active site of AMP kinase 1 indicated that the borano group may prevent two conserved critical Arg interactions with the alpha-phosphate, resulting in substrate bad positioning.
Collapse
Affiliation(s)
- D Topalis
- Laboratoire d'Enzymologie, Université Paris, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gondeau C, Chaloin L, Lallemand P, Roy B, Périgaud C, Barman T, Varga A, Vas M, Lionne C, Arold ST. Molecular basis for the lack of enantioselectivity of human 3-phosphoglycerate kinase. Nucleic Acids Res 2008; 36:3620-9. [PMID: 18463139 PMCID: PMC2441801 DOI: 10.1093/nar/gkn212] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Non-natural l-nucleoside analogues are increasingly used as therapeutic agents to treat cancer and viral infections. To be active, l-nucleosides need to be phosphorylated to their respective triphosphate metabolites. This stepwise phosphorylation relies on human enzymes capable of processing l-nucleoside enantiomers. We used crystallographic analysis to reveal the molecular basis for the low enantioselectivity and the broad specificity of human 3-phosphoglycerate kinase (hPGK), an enzyme responsible for the last step of phosphorylation of many nucleotide derivatives. Based on structures of hPGK in the absence of nucleotides, and bound to l and d forms of MgADP and MgCDP, we show that a non-specific hydrophobic clamp to the nucleotide base, as well as a water-filled cavity behind it, allows high flexibility in the interaction between PGK and the bases. This, combined with the dispensability of hydrogen bonds to the sugar moiety, and ionic interactions with the phosphate groups, results in the positioning of different nucleotides so to expose their diphosphate group in a position competent for catalysis. Since the third phosphorylation step is often rate limiting, our results are expected to alleviate in silico tailoring of l-type prodrugs to assure their efficient metabolic processing.
Collapse
Affiliation(s)
- C Gondeau
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, UMR 5236, CNRS-Universités Montpellier 1 et 2, Institut de Biologie, 4 bd Henri IV, CS69033, 34965 Montpellier cedex 2, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Topalis D, Kumamoto H, Alexandre JAC, Dugué L, Pochet S, Berteina-Raboin S, Agrofoglio LA, Deville-Bonne D. Looking for new pyrimidine acyclic nucleotide analogues designed for phosphorylation by human UMP-CMP kinase. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2008; 26:1369-73. [PMID: 18066785 DOI: 10.1080/15257770701533982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Human UMP-CMP kinase is involved in the phosphorylation of nucleic acid precursors and also in the activation of antiviral analogues including cidofovir, an acyclic phosphonate compound that mimicks dCMP and shows a broad antiviral spectrum. The binding of ligands to the enzyme was here investigated using a fluorescent probe and a competitive titration assay. At the acceptor site, the enzyme was found to accommodate any base, purine and pyrimidine, including thymidine. A method for screening analogues based on their affinity for the UMP binding site was developed. The affinities of uracil vinylphosphonate derivatives modified in the 5 position were found similar to (d)UMP and (d)CMP and improved when compared to cidofovir.
Collapse
Affiliation(s)
- Dimitri Topalis
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, CNRS-Université Paris 6, 4 place Jussieu, F-75005 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Alexandre JAC, Roy B, Topalis D, Périgaud C, Deville-Bonne D. Enantio-selectivity of human nucleoside monophosphate kinases. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2008; 26:1375-9. [PMID: 18066786 DOI: 10.1080/15257770701534014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Over recent years, there has been a renewed interest in the development of L-nucleosides as safe and efficacious drugs for the treatment of viral infections. Biological activity of these compounds requires phosphorylation to their triphosphate form, involving nucleoside monophosphate kinases in the second step. In order to characterize the activation pathway of L-nucleosides of the pyrimidine series, we studied the enantio-selectivity of human uridylate-cytidylate and thymidylate kinases. The results showed that these enzymes are only weakly enantio-selective and are thus probably involved in the activation of L-nucleosides in vivo. An activation pathway for telbivudine (L-dT) was therefore proposed.
Collapse
Affiliation(s)
- J A C Alexandre
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE2852-CNRS-Université Paris 6, 4 place Jussieu, F-75005 Paris, France
| | | | | | | | | |
Collapse
|
27
|
Alexandre JA, Roy B, Topalis D, Pochet S, Périgaud C, Deville-Bonne D. Enantioselectivity of human AMP, dTMP and UMP-CMP kinases. Nucleic Acids Res 2007; 35:4895-904. [PMID: 17626051 PMCID: PMC1950558 DOI: 10.1093/nar/gkm479] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
l-Nucleoside analogues such as lamivudine are active for treating viral infections. Like d-nucleosides, the biological activity of the l-enantiomers requires their stepwise phosphorylation by cellular or viral kinases to give the triphosphate. The enantioselectivity of NMP kinases has not been thoroughly studied, unlike that of deoxyribonucleoside kinases. We have therefore investigated the capacity of l-enantiomers of some natural (d)NMP to act as substrates for the recombinant forms of human uridylate-cytidylate kinase, thymidylate kinase and adenylate kinases 1 and 2. Both cytosolic and mitochondrial adenylate kinases were strictly enantioselective, as they phosphorylated only d-(d)AMP. l-dTMP was a substrate for thymidylate kinase, but with an efficiency 150-fold less than d-dTMP. Both l-dUMP and l-(d)CMP were phosphorylated by UMP-CMP kinase although much less efficiently than their natural counterparts. The stereopreference was conserved with the 2′-azido derivatives of dUMP and dUMP while, unexpectedly, the 2′-azido-d-dCMP was a 4-fold better substrate for UMP-CMP kinase than was CMP. Docking simulations showed that the small differences in the binding of d-(d)NMP to their respective kinases could account for the differences in interactions of the l-isomers with the enzymes. This in vitro information was then used to develop the in vivo activation pathway for l-dT.
Collapse
Affiliation(s)
- Julie A.C. Alexandre
- Laboratoire d’Enzymologie Moléculaire, FRE 2852-CNRS-Université Paris 6, 4, place Jussieu, 75005 Paris Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Universités Montpellier 1 et 2, case courrier 1705, Bâtiment Chimie 17, Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5 and Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex15, France
| | - Béatrice Roy
- Laboratoire d’Enzymologie Moléculaire, FRE 2852-CNRS-Université Paris 6, 4, place Jussieu, 75005 Paris Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Universités Montpellier 1 et 2, case courrier 1705, Bâtiment Chimie 17, Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5 and Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex15, France
| | - Dimitri Topalis
- Laboratoire d’Enzymologie Moléculaire, FRE 2852-CNRS-Université Paris 6, 4, place Jussieu, 75005 Paris Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Universités Montpellier 1 et 2, case courrier 1705, Bâtiment Chimie 17, Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5 and Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex15, France
| | - Sylvie Pochet
- Laboratoire d’Enzymologie Moléculaire, FRE 2852-CNRS-Université Paris 6, 4, place Jussieu, 75005 Paris Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Universités Montpellier 1 et 2, case courrier 1705, Bâtiment Chimie 17, Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5 and Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex15, France
| | - Christian Périgaud
- Laboratoire d’Enzymologie Moléculaire, FRE 2852-CNRS-Université Paris 6, 4, place Jussieu, 75005 Paris Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Universités Montpellier 1 et 2, case courrier 1705, Bâtiment Chimie 17, Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5 and Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex15, France
| | - Dominique Deville-Bonne
- Laboratoire d’Enzymologie Moléculaire, FRE 2852-CNRS-Université Paris 6, 4, place Jussieu, 75005 Paris Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Universités Montpellier 1 et 2, case courrier 1705, Bâtiment Chimie 17, Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5 and Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex15, France
- *To whom correspondence should be addressed.+33 1 44 27 59 93, Fax: +33 1 44 27 59 94
| |
Collapse
|
28
|
Topalis D, Kumamoto H, Amaya Velasco MF, Dugué L, Haouz A, Alexandre JAC, Gallois-Montbrun S, Alzari PM, Pochet S, Agrofoglio LA, Deville-Bonne D. Nucleotide binding to human UMP-CMP kinase using fluorescent derivatives -- a screening based on affinity for the UMP-CMP binding site. FEBS J 2007; 274:3704-3714. [PMID: 17608725 DOI: 10.1111/j.1742-4658.2007.05902.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Methylanthraniloyl derivatives of ATP and CDP were used in vitro as fluorescent probes for the donor-binding and acceptor-binding sites of human UMP-CMP kinase, a nucleoside salvage pathway kinase. Like all NMP kinases, UMP-CMP kinase binds the phosphodonor, usually ATP, and the NMP at different binding sites. The reaction results from an in-line phosphotransfer from the donor to the acceptor. The probe for the donor site was displaced by the bisubstrate analogs of the Ap5X series (where X = U, dT, A, G), indicating the broad specificity of the acceptor site. Both CMP and dCMP were competitors for the acceptor site probe. To find antimetabolites for antivirus and anticancer therapies, we have developed a method of screening acyclic phosphonate analogs that is based on the affinity of the acceptor-binding site of the human UMP-CMP kinase. Several uracil vinylphosphonate derivatives had affinities for human UMP-CMP kinase similar to those of dUMP and dCMP and better than that of cidofovir, an acyclic nucleoside phosphonate with a broad spectrum of antiviral activities. The uracil derivatives were inhibitors rather than substrates of human UMP-CMP kinase. Also, the 5-halogen-substituted analogs inhibited the human TMP kinase less efficiently. The broad specificity of the enzyme acceptor-binding site is in agreement with a large substrate-binding pocket, as shown by the 2.1 A crystal structure.
Collapse
Affiliation(s)
- Dimitri Topalis
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Hiroki Kumamoto
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Maria-Fernanda Amaya Velasco
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Laurence Dugué
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Ahmed Haouz
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Julie Anne C Alexandre
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Sarah Gallois-Montbrun
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Pedro Maria Alzari
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Sylvie Pochet
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Luigi André Agrofoglio
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Dominique Deville-Bonne
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS-Paris 6, Institut Jacques Monod, Paris, France Institut de Chimie Organique et Analytique, UMR CNRS 6005, FR 2708, Université d'Orléans, UFR Sciences, Orléans, France Unité de Biochimie Structurale, URA CNRS 2185, Institut Pasteur, Paris, France Unité de Chimie Organique, URA CNRS 2128, Institut Pasteur, Paris, France Plate-Forme 6- Cristallogénèse et Diffraction des Rayons X, Institut Pasteur, Paris, France Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, Paris, France
| |
Collapse
|
29
|
Balestri F, Barsotti C, Lutzemberger L, Camici M, Ipata PL. Key role of uridine kinase and uridine phosphorylase in the homeostatic regulation of purine and pyrimidine salvage in brain. Neurochem Int 2007; 51:517-23. [PMID: 17643556 DOI: 10.1016/j.neuint.2007.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 06/12/2007] [Accepted: 06/14/2007] [Indexed: 10/23/2022]
Abstract
Uridine, the major circulating pyrimidine nucleoside, participating in the regulation of a number of physiological processes, is readily uptaken into mammalian cells. The balance between anabolism and catabolism of intracellular uridine is maintained by uridine kinase, catalyzing the first step of UTP and CTP salvage synthesis, and uridine phosphorylase, catalyzing the first step of uridine degradation to beta-alanine in liver. In the present study we report that the two enzymes have an additional role in the homeostatic regulation of purine and pyrimidine metabolism in brain, which relies on the salvage synthesis of nucleotides from preformed nucleosides and nucleobases, rather than on the de novo synthesis from simple precursors. The experiments were performed in rat brain extracts and cultured human astrocytoma cells. The rationale of the reciprocal regulation of purine and pyrimidine salvage synthesis in brain stands (i) on the inhibition exerted by UTP and CTP, the final products of the pyrimidine salvage pathway, on uridine kinase and (ii) on the widely accepted idea that pyrimidine salvage occurs at the nucleoside level (mostly uridine), while purine salvage is a 5-phosphoribosyl-1-pyrophosphate (PRPP)-mediated process, occurring at the nucleobase level. Thus, at relatively low UTP and CTP level, uptaken uridine is mainly anabolized to uridine nucleotides. On the contrary, at relatively high UTP and CTP levels the inhibition of uridine kinase channels uridine towards phosphorolysis. The ribose-1-phosphate is then transformed into PRPP, which is used for purine salvage synthesis.
Collapse
Affiliation(s)
- Francesco Balestri
- Dipartimento di Biologia, Unità di Biochimica, Università di Pisa, Via S. Zeno 51, 56100 Pisa, Italy
| | | | | | | | | |
Collapse
|
30
|
Ofiteru A, Bucurenci N, Alexov E, Bertrand T, Briozzo P, Munier-Lehmann H, Gilles AM. Structural and functional consequences of single amino acid substitutions in the pyrimidine base binding pocket of Escherichia coli CMP kinase. FEBS J 2007; 274:3363-73. [PMID: 17542990 DOI: 10.1111/j.1742-4658.2007.05870.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bacterial CMP kinases are specific for CMP and dCMP, whereas the related eukaryotic NMP kinase phosphorylates CMP and UMP with similar efficiency. To explain these differences in structural terms, we investigated the contribution of four key amino acids interacting with the pyrimidine ring of CMP (Ser36, Asp132, Arg110 and Arg188) to the stability, catalysis and substrate specificity of Escherichia coli CMP kinase. In contrast to eukaryotic UMP/CMP kinases, which interact with the nucleobase via one or two water molecules, bacterial CMP kinase has a narrower NMP-binding pocket and a hydrogen-bonding network involving the pyrimidine moiety specific for the cytosine nucleobase. The side chains of Arg110 and Ser36 cannot establish hydrogen bonds with UMP, and their substitution by hydrophobic amino acids simultaneously affects the K(m) of CMP/dCMP and the k(cat) value. Substitution of Ser for Asp132 results in a moderate decrease in stability without significant changes in K(m) value for CMP and dCMP. Replacement of Arg188 with Met does not affect enzyme stability but dramatically decreases the k(cat)/K(m) ratio compared with wild-type enzyme. This effect might be explained by opening of the enzyme/nucleotide complex, so that the sugar no longer interacts with Asp185. The reaction rate for different modified CMP kinases with ATP as a variable substrate indicated that none of changes induced by these amino acid substitutions was 'propagated' to the ATP subsite. This 'modular' behavior of E. coli CMP kinase is unique in comparison with other NMP kinases.
Collapse
Affiliation(s)
- Augustin Ofiteru
- Laboratory of Enzymology and Applied Microbiology, Cantacuzino Institute, Bucharest, Romania
| | | | | | | | | | | | | |
Collapse
|
31
|
Sabini E, Hazra S, Konrad M, Lavie A. Nonenantioselectivity property of human deoxycytidine kinase explained by structures of the enzyme in complex with L- and D-nucleosides. J Med Chem 2007; 50:3004-14. [PMID: 17530837 PMCID: PMC2586175 DOI: 10.1021/jm0700215] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Biological molecules are predominantly enantioselective. Yet currently two nucleoside analogue prodrugs (3TC and FTC) with opposite chirality compared to physiological nucleosides are clinically approved for the treatment of HIV infections. These prodrugs require conversion to their triphosphorylated forms to achieve pharmacological activity. The first step in the activation of these agents is catalyzed by human deoxycytidine kinase (dCK). This enzyme possesses the ability to phosphorylate nucleosides of the unnatural L-chirality. To understand the molecular basis for the nonenantioselectivity of dCK, we solved the crystal structures of the enzyme in complex with the L-enantiomer and of its physiological substrate deoxycytidine and with the L-nucleoside analogue FTC. These were compared to a structure solved with D-dC. Our results highlight structural adjustments imposed on the L-nucleosides and properties of the enzyme endowing it with the ability to phosphorylate substrates with nonphysiological chirality. This work reveals the molecular basis for the activation of L-nucleosides by dCK.
Collapse
Affiliation(s)
- Elisabetta Sabini
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S. Ashland (M/C 669), Chicago, IL 60607, USA
| | - Saugata Hazra
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S. Ashland (M/C 669), Chicago, IL 60607, USA
| | - Manfred Konrad
- Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077 Göttingen, Germany
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S. Ashland (M/C 669), Chicago, IL 60607, USA
| |
Collapse
|
32
|
Topalis D, Collinet B, Gasse C, Dugué L, Balzarini J, Pochet S, Deville-Bonne D. Substrate specificity of vaccinia virus thymidylate kinase. FEBS J 2006; 272:6254-65. [PMID: 16336263 DOI: 10.1111/j.1742-4658.2005.05006.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Anti-poxvirus therapies are currently limited to cidofovir [(S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine], but drug-resistant strains have already been characterized. In the aim of finding a new target, the thymidylate (TMP) kinase from vaccinia virus, the prototype of Orthopoxvirus, has been overexpressed in Escherichia coli after cloning the gene (A48R). Specific inhibitors and alternative substrates of pox TMP kinase should contribute to virus replication inhibition. Biochemical characterization of the enzyme revealed distinct catalytic features when compared to its human counterpart. Sharing 42% identity with human TMP kinase, the vaccinia virus enzyme was assumed to adopt the common fold of nucleoside monophosphate kinases. The enzyme was purified to homogeneity and behaves as a homodimer, like all known TMP kinases. Initial velocity studies showed that the Km for ATP-Mg2+ and dTMP were 0.15 mm and 20 microM, respectively. Vaccinia virus TMP kinase was found to phosphorylate dTMP, dUMP and also dGMP from any purine and pyrimidine nucleoside triphosphate. 5-Halogenated dUMP such as 5-iodo-2'-deoxyuridine 5'-monophosphate (5I-dUMP) and 5-bromo-2'-deoxyuridine 5'-monophosphate (5Br-dUMP) were also efficient alternative substrates. Using thymidine-5'-(4-N'-methylanthraniloyl-aminobutyl)phosphoramidate as a fluorescent probe of the dTMP binding site, we detected an ADP-induced conformational change enhancing the binding affinity of dTMP and analogues. Several thymidine and dTMP derivatives were found to bind the enzyme with micromolar affinities. The present study provides the basis for the design of specific inhibitors or substrates for poxvirus TMP kinase.
Collapse
Affiliation(s)
- Dimitri Topalis
- Laboratoire d'Enzymologie Moléculaire et Fonctionnelle, FRE 2852 CNRS, Paris, France
| | | | | | | | | | | | | |
Collapse
|
33
|
Gallois-Montbrun S, Faraj A, Seclaman E, Sommadossi JP, Deville-Bonne D, Véron M. Broad specificity of human phosphoglycerate kinase for antiviral nucleoside analogs. Biochem Pharmacol 2004; 68:1749-56. [PMID: 15450940 DOI: 10.1016/j.bcp.2004.06.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Accepted: 06/14/2004] [Indexed: 10/26/2022]
Abstract
Nucleoside analogs used in antiviral therapies need to be phosphorylated to their tri-phospho counterparts in order to be active on their cellular target. Human phosphoglycerate kinase (hPGK) was recently reported to participate in the last step of phosphorylation of cytidine L-nucleotide derivatives [Krishnan PGE, Lam W, Dutschman GE, Grill SP, Cheng YC. Novel role of 3-phosphoglycerate kinase, a glycolytic enzyme, in the activation of L-nucleoside analogs, a new class of anticancer and antiviral agents. J Biol Chem 2003;278:36726-32]. In the present work, we extended the enzymatic study of human PGK specificity to purine and pyrimidine nucleotide derivatives in both D- and L-configuration. Human PGK demonstrated catalytic efficiencies in the 10(4)-10(5)M(-1)s(-1) range for purine ribo-, deoxyribo- and dideoxyribonucleotide derivatives, either in D- or L-configuration. In contrast, it was poorly active with natural pyrimidine D-nucleotides (less than 10(3)M(-1)s(-1)). Pyrimidine L-enantiomers, which are promising therapeutic analogs against B hepatitis, were 2-25 times better substrates than their D-counterparts. The broad specificity of substrate of human PGK suggests that this enzyme may be involved in the cellular activation of several antiviral nucleoside analogs including dideoxyinosine, acyclovir, L-2'-deoxycytosine and L-2'-deoxythymidine.
Collapse
Affiliation(s)
- Sarah Gallois-Montbrun
- Unité de Régulation Enzymatique des Activités Cellulaires, CNRS URA 2185, Institut Pasteur, 75724 Paris Cedex 15, France
| | | | | | | | | | | |
Collapse
|
34
|
Hsu CH, Liou JY, Dutschman GE, Cheng YC. Phosphorylation of Cytidine, Deoxycytidine, and Their Analog Monophosphates by Human UMP/CMP Kinase Is Differentially Regulated by ATP and Magnesium. Mol Pharmacol 2004; 67:806-14. [PMID: 15550676 DOI: 10.1124/mol.104.006098] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human UMP/CMP kinase (cytidylate kinase; EC 2.7.4.14) is responsible for phosphorylation of CMP, UMP, and deoxycytidine monophosphate (dCMP) and also plays an important role in the activation of pyrimidine analogs, some of which are clinically useful anticancer or antiviral drugs. Previous kinetic data using recombinant or highly purified human UMP/CMP kinase showed that dCMP, as well as pyrimidine analog monophosphates, were much poorer substrates than CMP or UMP for this enzyme. This implies that other unidentified mechanisms must be involved to make phosphorylation of dCMP or pyrimidine analog monophosphates inside cells by this enzyme possible. Here, we reevaluated the optimal reaction conditions for human recombinant human UMP/CMP kinase to phosphorylate dCMP and CMP (referred as dCMPK and CMPK activities). We found that ATP and magnesium were important regulators of the kinase activities of this enzyme. Free magnesium enhanced dCMPK activity but inhibited CMPK activity. Free ATP or excess ATP/magnesium, on the other hand, inhibited dCMPK but not CMPK reactions. The differential regulation of dCMPK versus CMPK activities by ATP or magnesium was also seen in other 2'-deoxypyrimidine analog monophosphates (deoxyuridine monophosphate, 5-fluorodeoxyuridine monophosphate, 1-beta-D-arabinofuranosylcytosine monophosphate, and gemcitabine monophosphate) versus their ribose-counterparts (UMP and 5-fluorouridine monophosphate), in a similar manner. The data suggest that the active sites of human UMP/CMP kinase for dCMP and for CMP cannot be identical. Furthermore, enzyme inhibition studies demonstrated that CMP could inhibit dCMP phosphorylation in a noncompetitive manner, with Ki values much higher than its own Km values. We thus propose novel models for the phosphorylation action of human UMP/CMP kinase.
Collapse
Affiliation(s)
- Chih-Hung Hsu
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar St., SHM B226, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
35
|
Roy B, Verri A, Lossani A, Spadari S, Focher F, Aubertin AM, Gosselin G, Mathé C, Périgaud C. Enantioselectivity of ribonucleotide reductase: a first study using stereoisomers of pyrimidine 2′-azido-2′-deoxynucleosides. Biochem Pharmacol 2004; 68:711-8. [PMID: 15276078 DOI: 10.1016/j.bcp.2004.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2003] [Accepted: 05/06/2004] [Indexed: 11/16/2022]
Abstract
In this paper, the enantioselectivity of ribonucleotide reductase (RNR, EC 1.17.4.1), a pivotal enzyme involved in DNA biosynthesis, was studied using the beta-d and beta-l stereoisomers of 2'-azido-2'-deoxynucleosides of uracil and cytosine. The corresponding 5'-diphosphate derivatives in the d-configuration have been extensively studied as mechanism-based inhibitors of the enzyme. The original l-enantiomers were synthesized and evaluated in vitro. In cell culture experiments, only the cytosine derivative with a d-configuration was found cytostatic and able to deplete dNTP pools in response to RNR inhibition. In the case of the uracil enantiomeric pair, this result correlates with an inefficient intracellular monophosphorylation as demonstrated in testing their substrate properties against human uridine-cytidine kinase 1. Regarding cytosine analogues, human deoxycytidine kinase was found to be able to phosphorylate both enantiomers with comparable efficiency but only the d-stereoisomer was active in human cell culture. The interaction of the beta-d and beta-l stereoisomers of 2'-azido-2'-deoxyuridine 5'-diphosphate with purified Escherichia coli RNR was also examined. Inactivation of the enzyme was only observed in the presence of the d-stereoisomer, demonstrating that RNR exhibits enantiospecificity with respect to the natural configuration of the sugar moiety, as far as 2'-azido-2'-deoxynucleotides are concerned.
Collapse
Affiliation(s)
- Béatrice Roy
- UMR 5625 CNRS-UM II, Université Montpellier II, case courrier 008, place E. Bataillon, 34095 Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Segura-Peña D, Sekulic N, Ort S, Konrad M, Lavie A. Substrate-induced conformational changes in human UMP/CMP kinase. J Biol Chem 2004; 279:33882-9. [PMID: 15163660 DOI: 10.1074/jbc.m401989200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human UMP/CMP kinase plays a crucial role in supplying precursors for nucleic acid synthesis by catalyzing the conversion of UMP, CMP, and dCMP into their diphosphate form. In addition, this kinase is an essential component of the activation cascade of medicinally relevant nucleoside analog prodrugs such as AraC, gemcitabine, and ddC. During the catalytic cycle the enzyme undergoes large conformational changes from open in the absence of substrates to closed in the presence of both phosphoryl donor and phosphoryl acceptor. Here we report the crystal structure of the substrate-free, open form of human UMP/CMP kinase. Comparison of the open structure with the closed state previously reported for the similar Dictyostelium discoideum UMP/CMP kinase reveals the conformational changes that occur upon substrate binding. We observe a classic example of induced fit where substrate-induced conformational changes in hinge residues result in rigid body movements of functional domains to form the catalytically competent state. In addition, a homology model of the human enzyme in the closed state based on the structure of D. discoideum UMP/CMP kinase aids to rationalize the substrate specificity of the human enzyme.
Collapse
Affiliation(s)
- Dario Segura-Peña
- University of Illinois at Chicago, Department of Biochemistry and Molecular Genetics, Chicago, Illinois 60607, USA
| | | | | | | | | |
Collapse
|
37
|
Liu M, Cheng J, Zhang SL, Wang L, Shao Q, Zhang J, Liang YD. Screening of HCTP4 interacting proteins in leukocytes by yeast-two hybrid technique. Shijie Huaren Xiaohua Zazhi 2004; 12:832-835. [DOI: 10.11569/wcjd.v12.i4.832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the biological function of HCTP4, yeast-two hybrid was performed to screen proteins interacting with HCTP4 in leukocytes.
METHODS: The HCTP4 gene was amplified by polymerase chain reaction (PCR) and HCTP4 bait plasmid was constructed by using yeast-two hybrid system 3, then the constructed vector was transformed into yeast AH109. The transformed yeast mated with yeast Y187 containing leukocytes cDNA library plasmid in 2×YPDA medium. Diploid yeast was plated on synthetic dropout nutrient medium (SD/-Trp-Leu-His-Ade) and synthetic dropout nutrient medium (SD/-Trp-Leu-His-Ade) containing x--gal for selecting two times and screening. After extracting and sequencing of plasmid from blue colonies, we underwent analysis by bioinformatics.
RESULTS: Forty-four colonies were sequenced, among which twenty-five colonies were immunoglobulin lambda light chain, six human DNA sequences from clone RP11-189K21, four human DNA sequences from clone RP11-507C10, two homo sapiens 12p BAC RPCI11-75L1, one homo sapiens BAC clone RP11-21M10, one homo sapiens ubiquitin ligase mind bomb (MIB), one homo sapiens genomic DNA, chromosome 11 clone: RP11-867O8, one human DNA sequence from clone RP3-509I19, one homo sapiens small nuclear ribonucleoprotein polypeptide G, one homo sapiens UMP-CMP kinase (UMP-CMPK), and a new gene.
CONCLUSION: Genes of HCTP4 interacting proteins in leukocytes are successfully cloned and the results bring some new clues for studying the biological functions of HCTP4 and associated proteins.
Collapse
|