Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters

Publication year range
1.
Nature ; 558(7711): 610-614, 2018 06.
Article in English | MEDLINE | ID: mdl-29925952

ABSTRACT

Viral infections continue to represent major challenges to public health, and an enhanced mechanistic understanding of the processes that contribute to viral life cycles is necessary for the development of new therapeutic strategies 1 . Viperin, a member of the radical S-adenosyl-L-methionine (SAM) superfamily of enzymes, is an interferon-inducible protein implicated in the inhibition of replication of a broad range of RNA and DNA viruses, including dengue virus, West Nile virus, hepatitis C virus, influenza A virus, rabies virus 2 and HIV3,4. Viperin has been suggested to elicit these broad antiviral activities through interactions with a large number of functionally unrelated host and viral proteins3,4. Here we demonstrate that viperin catalyses the conversion of cytidine triphosphate (CTP) to 3'-deoxy-3',4'-didehydro-CTP (ddhCTP), a previously undescribed biologically relevant molecule, via a SAM-dependent radical mechanism. We show that mammalian cells expressing viperin and macrophages stimulated with IFNα produce substantial quantities of ddhCTP. We also establish that ddhCTP acts as a chain terminator for the RNA-dependent RNA polymerases from multiple members of the Flavivirus genus, and show that ddhCTP directly inhibits replication of Zika virus in vivo. These findings suggest a partially unifying mechanism for the broad antiviral effects of viperin that is based on the intrinsic enzymatic properties of the protein and involves the generation of a naturally occurring replication-chain terminator encoded by mammalian genomes.


Subject(s)
Antiviral Agents/metabolism , Cytidine Triphosphate/metabolism , Genome, Human/genetics , Proteins/genetics , Proteins/metabolism , Transcription Termination, Genetic , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Cytidine Triphosphate/biosynthesis , Cytidine Triphosphate/chemistry , HEK293 Cells , Humans , Oxidoreductases Acting on CH-CH Group Donors , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Ribonucleotides , Substrate Specificity , Vero Cells , Zika Virus/enzymology , Zika Virus/metabolism
2.
J Biol Chem ; 294(17): 6888-6898, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30872404

ABSTRACT

Virus-inhibitory protein, endoplasmic reticulum-associated, interferon-inducible (viperin) is a radical SAM enzyme that plays a multifaceted role in the cellular antiviral response. Viperin has recently been shown to catalyze the SAM-dependent formation of 3'-deoxy-3',4'-didehydro-CTP (ddhCTP), which inhibits some viral RNA polymerases. Viperin is also implicated in regulating Lys-63-linked polyubiquitination of interleukin-1 receptor-associated kinase-1 (IRAK1) by the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6) as part of the Toll-like receptor-7 and -9 (TLR7/9) innate immune signaling pathways. In these pathways, the poly-ubiquitination of IRAK1 by TRAF6 is necessary to activate IRAK1, which then phosphorylates downstream targets and ultimately leads to the production of type I interferons. That viperin is a component of these pathways suggested that its enzymatic activity might be regulated by interactions with partner proteins. To test this idea, we have reconstituted the interactions between viperin, IRAK1, and TRAF6 by transiently expressing these enzymes in HEK 293T cells. We show that IRAK1 and TRAF6 increase viperin activity ∼10-fold to efficiently catalyze the radical-mediated dehydration of CTP to ddhCTP. Furthermore, we found that TRAF6-mediated ubiquitination of IRAK1 requires the association of viperin with both IRAK1 and TRAF6. Ubiquitination appears to depend on structural changes in viperin induced by SAM binding, but, significantly, does not require catalytically active viperin. We conclude that the synergistic activation of viperin and IRAK1 provides a mechanism that couples innate immune signaling with the production of the antiviral nucleotide ddhCTP.


Subject(s)
Antiviral Agents/metabolism , Cytidine Triphosphate/biosynthesis , Immunity, Innate , Interleukin-1 Receptor-Associated Kinases/metabolism , Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Adenosine/administration & dosage , Adenosine/analogs & derivatives , HEK293 Cells , Half-Life , Humans , Intracellular Signaling Peptides and Proteins , Oxidoreductases Acting on CH-CH Group Donors , Phosphorylation , Protein Binding , S-Adenosylmethionine/metabolism , Ubiquitination
3.
Antimicrob Agents Chemother ; 59(10): 6328-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26248377

ABSTRACT

The prevention of mother-to-child transmission (MTCT) of HIV is a crucial component in HIV therapy. Nucleoside reverse transcriptase inhibitors (NRTIs), primarily 3'-azido-3'-thymidine (AZT [zidovudine]), have been used to treat both mothers and neonates. While AZT is being replaced with less toxic drugs in treating mothers in MTCT prevention, it is still commonly used to treat neonates. Problems related to mitochondrial toxicity and potential mutagenesis associated with AZT treatment have been reported in treated cohorts. Yet little is known concerning the metabolism and potential toxicity of AZT on embryonic and neonatal tissues, especially considering that the enzymes of nucleoside metabolism change dramatically as many tissues convert from hyperplastic to hypertrophic growth during this period. AZT is known to inhibit thymidine phosphorylation and potentially alter deoxynucleoside triphosphate (dNTP) pools in adults. This study examines the effects of AZT on dNTP pools, mRNA expression of deoxynucleoside/deoxynucleotide metabolic enzymes, and mitochondrial DNA levels in a neonatal rat model. Results show that AZT treatment dramatically altered dNTP pools in the first 7 days of life after birth, which normalized to age-matched controls in the second and third weeks. Additionally, AZT treatment dramatically increased the mRNA levels of many enzymes involved in deoxynucleotide synthesis and mitochondrial biogenesis during the first week of life, which normalized to age-matched controls by the third week. These results were correlated with depletion of mitochondrial DNA noted in the second week. Taken together, results demonstrated that AZT treatment has a powerful effect on the deoxynucleotide synthesis pathways that may be associated with toxicity and mutagenesis.


Subject(s)
Anti-HIV Agents/toxicity , DNA, Mitochondrial/antagonists & inhibitors , Heart/drug effects , RNA, Messenger/antagonists & inhibitors , Reverse Transcriptase Inhibitors/toxicity , Zidovudine/toxicity , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/biosynthesis , Animals , Animals, Newborn , Cytidine Triphosphate/antagonists & inhibitors , Cytidine Triphosphate/biosynthesis , DNA Copy Number Variations/drug effects , DNA, Mitochondrial/biosynthesis , Female , Gene Expression Regulation , Guanosine Triphosphate/antagonists & inhibitors , Guanosine Triphosphate/biosynthesis , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phosphorylation/drug effects , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Uridine Triphosphate/antagonists & inhibitors , Uridine Triphosphate/biosynthesis
4.
Int J Oncol ; 34(3): 813-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212686

ABSTRACT

Cyclopentenyl cytosine (CPEC), targetting the de novo biosynthesis of cytidine triphosphate (CTP), increases the cytotoxicity of gemcitabine (2',2'-difluoro-2'-deoxycytidine, dFdC) alone and in combination with irradiation in several human tumour cells in vitro. We investigated whether CPEC enhances the therapeutic ratio of gemcitabine and irradiation in human pancreatic BxPC-3 xenografts and in rat syngeneic L44 lung tumours. These models were selected because gemcitabine and radiation are used to treat both pancreatic and lung cancer patients and both models differ in growth capacity and in gemcitabine-induced radiosensitisation. A profound dose-dependent CTP-depletion was observed after a single injection of CPEC in both tumour tissue and in normal jejunum. In both models, CPEC alone induced a slight but significant tumour growth delay. The combination of CPEC with gemcitabine, at time intervals that showed CTP-depletion after CPEC, enhanced neither tumour growth delay nor toxicity as compared to gemcitabine alone. In addition, no beneficial effect of CPEC was observed in combination with gemcitabine and radiation. These results suggest that CPEC and gemcitabine alone as well as in combination with radiation target a similar cell population in both tumour models. In conclusion, future clinical development of CPEC as a modulator of gemcitabine combined with radiation is unlikely.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cytidine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Cytidine/pharmacology , Cytidine Triphosphate/biosynthesis , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Synergism , Female , Humans , Mice , Mice, Nude , Middle Aged , Rats , Rats, Inbred BN , Gemcitabine
5.
FASEB J ; 22(11): 3938-46, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18606862

ABSTRACT

This study examined the effects on cognitive behaviors of giving normal adult gerbils three compounds, normally in the circulation, which interact to increase brain phosphatides, synaptic proteins, dendritic spines, and neurotransmitter release. Animals received supplemental uridine (as its monophosphate, UMP; 0.5%) and choline (0.1%) via the diet, and docosahexaenoic acid (DHA; 300 mg/kg/day) by gavage, for 4 wk, and then throughout the subsequent period of behavioral training and testing. As shown previously, giving all three compounds caused highly significant (P<0.001) increases in total brain phospholipids and in each major phosphatide; giving DHA or UMP (plus choline) produced smaller increases in some of the phosphatides. DHA plus choline improved performance on the four-arm radial maze, T-maze, and Y-maze tests; coadministering UMP further enhanced these increases. (Uridine probably acts by generating both CTP, which can be limiting in phosphatide synthesis, and UTP, which activates P2Y receptors coupled to neurite outgrowth and protein synthesis. All three compounds also act by enhancing the substrate-saturation of phosphatide-synthesizing enzymes.) These findings demonstrate that a treatment that increases synaptic membrane content can enhance cognitive functions in normal animals.


Subject(s)
Brain Chemistry/drug effects , Diet , Docosahexaenoic Acids/pharmacology , Maze Learning/drug effects , Nerve Tissue Proteins/biosynthesis , Uridine Monophosphate/pharmacology , Animals , Behavior, Animal , Choline/pharmacology , Cytidine Triphosphate/biosynthesis , Gerbillinae , Lipotropic Agents/pharmacology , Male , Memory , Neurites/metabolism , Protein Biosynthesis/drug effects , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/metabolism , Synaptic Membranes/metabolism , Time Factors , Uridine Triphosphate/biosynthesis
6.
J Sep Sci ; 31(4): 689-95, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18307164

ABSTRACT

A novel separation method was developed to isolate directly cytidine triphosphate (CTP) from fermentation broth of yeast using anion-exchange supermacroporous cryogel. The anion-exchange cryogel with tertiary amine groups was prepared by graft polymerization. The breakthrough characteristics and elution performance of pure CTP in the cryogel bed were investigated experimentally and the CTP binding capacity was determined. Then the separation experiments of CTP from crude fermentation broth of yeast using the cryogel column were carried out using deionized water and 0.01 M HCl as washing buffer, respectively. The chromatographic behavior was monitored and analyzed. The purity and concentration of the obtained CTP in these processes were determined quantitatively by HPLC. The maximal purity of CTP obtained at the condition of 0.01 M HCl as washing buffer and 0.5 M NaCl in 0.01 M HCl as elution buffer reached 93%.


Subject(s)
Blood Proteins , Culture Media, Conditioned/chemistry , Cytidine Triphosphate/isolation & purification , Fermentation , Fibronectins , Saccharomyces cerevisiae/metabolism , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Cryogels , Cytidine Triphosphate/biosynthesis , Cytidine Triphosphate/chemistry , Hydrogels
8.
Appl Biochem Biotechnol ; 144(1): 1-14, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18415982

ABSTRACT

Conditions were studied in the biosynthesis of cytidine 5'-triphosphate (CTP) from cytidine 5'-monophosphate (CMP). A 201 x 7 anion ion-exchange resin was applied for the separation of CTP from CMP. Adsorption isotherm and elution conditions (eluant, eluant concentration, flow rate, sample volume loaded) were investigated. At the same time, a new high-performance liquid chromatography on an anion ion-exchange column WAX-1 with UV detector at 260 nm was developed to measure CMP, cytidine 5'-diphosphate (CDP), and CTP. The retention time for CMP, CDP, and CTP are 0.723, 1.448, and 4.432 min, respectively. This new rapid high-performance liquid chromatography (HPLC) method for the analysis of cytidine compounds in biological sample has a wide linear range with high precision and repeatability.


Subject(s)
Chromatography, High Pressure Liquid/methods , Chromatography, Ion Exchange/methods , Cytidine Monophosphate/metabolism , Cytidine Triphosphate/biosynthesis , Cytidine Triphosphate/isolation & purification , Adsorption , Biotechnology , Cytidine Diphosphate/analysis , Cytidine Monophosphate/analysis , Cytidine Triphosphate/analysis , Ion Exchange Resins , Saccharomyces cerevisiae/metabolism
9.
Biochim Biophys Acta Gen Subj ; 1862(12): 2714-2727, 2018 12.
Article in English | MEDLINE | ID: mdl-30251661

ABSTRACT

Molecular gates within enzymes often play important roles in synchronizing catalytic events. We explored the role of a gate in cytidine-5'-triphosphate synthase (CTPS) from Escherichia coli. This glutamine amidotransferase catalyzes the biosynthesis of CTP from UTP using either l-glutamine or exogenous NH3 as a substrate. Glutamine is hydrolyzed in the glutaminase domain, with GTP acting as a positive allosteric effector, and the nascent NH3 passes through a gate located at the end of a ~25-Å tunnel before entering the synthase domain where CTP is generated. Substitution of the gate residue Val 60 by Ala, Cys, Asp, Trp, or Phe using site-directed mutagenesis and subsequent kinetic analyses revealed that V60-substitution impacts glutaminase activity, nucleotide binding, salt-dependent inhibition, and inter-domain NH3 transport. Surprisingly, the increase in steric bulk present in V60F perturbed the local structure consistent with "pinching" the tunnel, thereby revealing processes that synchronize the transfer of NH3 from the glutaminase domain to the synthase domain. V60F had a slightly reduced coupling efficiency at maximal glutaminase activity that was ameliorated by slowing down the glutamine hydrolysis reaction, consistent with a "bottleneck" effect. The inability of V60F to use exogenous NH3 was overcome in the presence of GTP, and more so if CTPS was covalently modified by 6-diazo-5-oxo-l-norleucine. Use of NH2OH by V60F as an alternative bulkier substrate occurred most efficiently when it was concomitant with the glutaminase reaction. Thus, the glutaminase activity and GTP-dependent activation act in concert to open the NH3 gate of CTPS to mediate inter-domain NH3 transport.


Subject(s)
Ammonia/metabolism , Carbon-Nitrogen Ligases/metabolism , Adenosine Triphosphate/metabolism , Alkylation , Allosteric Regulation , Amino Acid Sequence , Binding Sites , Carbon-Nitrogen Ligases/chemistry , Catalysis , Chlorides/metabolism , Crystallography, X-Ray , Cytidine Triphosphate/biosynthesis , Glutaminase/metabolism , Glutamine/chemistry , Glutamine/metabolism , Hydrolysis , Kinetics , Mutagenesis, Site-Directed , Protein Conformation , Uridine Triphosphate/metabolism , Valine/chemistry , Valine/genetics , Valine/metabolism
10.
Biochim Biophys Acta ; 1764(2): 199-210, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16427816

ABSTRACT

Cytidine 5'-triphosphate synthase (CTPS) catalyzes the ATP-dependent formation of CTP from UTP using either NH3 or L-glutamine as the source of nitrogen. To identify the location of the ATP-binding site within the primary structure of E. coli CTPS, we used the affinity label 2',3'-dialdehyde adenosine 5'-triphosphate (oATP). oATP irreversibly inactivated CTPS in a first-order, time-dependent manner while ATP protected the enzyme from inactivation. In the presence of 10 mM UTP, the values of k(inact) and K(I) were 0.054 +/- 0.001 min(-1) and 3.36 +/- 0.02 mM, respectively. CTPS was labeled using (2,8-3H)oATP and subsequently subjected to trypsin-catalyzed proteolysis. The tryptic peptides were separated using reversed-phase HPLC, and two peptides were identified using N-terminal sequencing (S(492)GDDQLVEIIEVPNH(506) and Y(298)IELPDAY(K(306)) in a 5:1 ratio). The latter suggested that Lys 306 had been modified by oATP. Replacement of Lys 306 by alanine reduced the rate of oATP-dependent inactivation (k(inact) = 0.0058 +/- 0.0005 min(-1), K(I) = 3.7 +/- 1.3 mM) and reduced the apparent affinity of CTPS for both ATP and UTP by approximately 2-fold. The efficiency of K306A-catalyzed glutamine-dependent CTP formation was also reduced 2-fold while near wild-type activity was observed when NH3 was the substrate. These findings suggest that Lys 306 is not essential for ATP binding, but does play a role in bringing about the conformational changes that mediate interactions between the ATP and UTP sites, and between the ATP-binding site and the glutamine amide transfer domain. Replacement of the nearby, fully conserved Lys 297 by alanine did not affect NH3-dependent CTP formation, relative to wild-type CTPS, but reduced k(cat) for the glutaminase activity 78-fold. Our findings suggest that the conformational change associated with binding ATP may be transmitted through the L10-alpha11 structural unit (residues 297-312) and thereby mediate effects on the glutaminase activity of CTPS.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Carbon-Nitrogen Ligases/antagonists & inhibitors , Carbon-Nitrogen Ligases/chemistry , Escherichia coli/enzymology , Lysine/chemistry , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Binding Sites , Carbon-Nitrogen Ligases/genetics , Cytidine Triphosphate/biosynthesis , DNA Mutational Analysis , Kinetics , Lysine/genetics , Molecular Sequence Data , Mutation , Protein Conformation
11.
FEBS J ; 274(8): 1983-90, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17355283

ABSTRACT

Mollicutes are wall-less bacteria and cause various diseases in humans, animals and plants. They have the smallest genomes with low G + C content and lack many genes of DNA, RNA and protein precursor biosynthesis. Nucleoside diphosphate kinase (NDK), a house-keeping enzyme that plays a critical role in the synthesis of nucleic acids precursors, i.e. NTPs and dNTPs, is absent in all the Mollicutes genomes sequenced to date. Therefore, it would be of interest to know how Mollicutes synthesize dNTPs/NTPs without NDK. To answer this question, nucleoside monophosphate kinases (NMPKs) from Ureaplasma were studied regarding their role in the synthesis of NTPs/dNTPs. In this work, Ureaplasma adenylate kinase, cytidylate kinase, uridylate kinase and thymidylate kinase were cloned and expressed in Escherichia coli. The recombinant enzymes were purified and characterized. These NMPKs are base specific, as indicated by their names, and capable of converting (d)NMPs directly to (d)NTPs. The catalytic rates of (d)NTPs and (d)NDP synthesis by these NMPKs were determined using tritium-labelled (d)NMPs, and the rates for (d)NDP synthesis, in general, were much higher (up to 100-fold) than that of (d)NTP. Equilibrium studies with adenylate kinase suggested that the rates of NTPs/dNTPs synthesis by NMPKs in vivo are probably regulated by the levels of (d)NMPs. These results strongly indicate that NMPKs could substitute the NDK function in vivo.


Subject(s)
Adenosine Triphosphate/biosynthesis , Cytidine Triphosphate/biosynthesis , Guanosine Triphosphate/biosynthesis , Nucleoside-Phosphate Kinase/physiology , Ureaplasma/enzymology , Adenylate Kinase/physiology , Cloning, Molecular , Nucleoside-Diphosphate Kinase/physiology , Substrate Specificity
12.
Cancer Res ; 51(9): 2291-5, 1991 May 01.
Article in English | MEDLINE | ID: mdl-1707749

ABSTRACT

(6R)-5,10-Dideaza-5,6,7,8-tetrahydrofolic acid [(6R)DDATHF] is a folate antimetabolite with activity specifically directed against de novo purine synthesis, primarily through inhibition of glycinamide ribonucleotide transformylase. This inhibition resulted in major changes in the size of the nucleotide pools in CCRF-CEM cells. After a 4-h incubation with 1 microM (6R)DDATHF, dramatic reductions in the ATP and GTP pools were observed, with almost no effect on CTP, UTP, and deoxyribonucleotide pools. When the incubation was continued in drug-free medium, recovery of ATP and GTP pools was protracted. ATP did not return to normal until 24-36 h, and GTP pools were only partially repleted by 48 h. The ATP and GTP pools were not affected when the initial 4-h incubation with (6R)DDATHF was conducted in the presence of 100 microM hypoxanthine. Addition of hypoxanthine to the medium after a 4-h incubation with (6R)DDATHF caused rapid recovery of the ATP and GTP pools. Similar effects were seen when the purine precursor aminoimidazole carboxamide was used in place of hypoxanthine. The effect of (6R)DDATHF on nucleotide pools and the capability of hypoxanthine or aminoimidazole carboxamide to prevent or reverse this phenomenon correlated directly with the inhibition of cell growth. Presumably as a consequence of the decrease in purine nucleotide triphosphate levels, the conversion of exogenously added uridine, thymidine, and deoxyuridine to nucleotides was markedly decreased. These effects were protracted for almost 48 h and were also reversed by hypoxanthine. Differential repletion of ATP and GTP pools after (6R)DDATHF pre-treatment demonstrated that diminished precursor phosphorylation is primarily a consequence of GTP rather than ATP starvation.


Subject(s)
Adenosine Triphosphate/biosynthesis , Folic Acid Antagonists/pharmacology , Guanosine Triphosphate/biosynthesis , Leukemia-Lymphoma, Adult T-Cell/metabolism , Tetrahydrofolates/pharmacology , Aminoimidazole Carboxamide/pharmacology , Cytidine Triphosphate/biosynthesis , DNA/biosynthesis , Humans , Hypoxanthine , Hypoxanthines/pharmacology , RNA/biosynthesis , Thymidylate Synthase/metabolism , Time Factors , Tumor Cells, Cultured/metabolism , Uridine Triphosphate/biosynthesis
13.
Oncotarget ; 7(9): 10386-401, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26824416

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.


Subject(s)
Cytosine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Nucleoside-Phosphate Kinase/metabolism , Organophosphonates/pharmacology , Papillomavirus Infections/drug therapy , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/virology , ATP-Binding Cassette Transporters/biosynthesis , Cell Line, Tumor , Cidofovir , Cytidine Triphosphate/biosynthesis , Cytosine/pharmacology , Female , HeLa Cells , Humans , Microbial Sensitivity Tests , Nucleoside-Phosphate Kinase/biosynthesis , Papillomaviridae , Phosphorylation , Solute Carrier Proteins/biosynthesis , Uridine Triphosphate/biosynthesis , Uterine Cervical Neoplasms/pathology
14.
Biochim Biophys Acta ; 1699(1-2): 213-20, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15158730

ABSTRACT

Cytidine 5'-triphosphate (CTP) synthase catalyzes the ATP-dependent formation of CTP from UTP using either ammonia or l-glutamine as the source of nitrogen. When glutamine is the substrate, GTP is required as a positive allosteric effector to promote catalysis of glutamine hydrolysis. We show that at concentrations exceeding approximately 0.15 mM, GTP actually behaves as a negative allosteric effector of E. coli CTP synthase, inhibiting glutamine-dependent CTP formation. In addition, GTP inhibits NH(3)-dependent CTP formation in a concentration-dependent manner. However, GTP does not inhibit the enzyme's intrinsic glutaminase activity. Although the activation of CTP synthase by GTP does not display cooperative behavior, inhibition of both CTP synthase-catalyzed ammonia- and glutamine-dependent CTP synthesis by GTP do exhibit positive cooperativity. These results suggest that GTP binding affects CTP synthase catalysis in two ways: it activates enzyme-catalyzed glutamine hydrolysis and it inhibits the utilization of NH(3) as a substrate by the synthase domain.


Subject(s)
Carbon-Nitrogen Ligases/antagonists & inhibitors , Cytidine Triphosphate/biosynthesis , Escherichia coli/enzymology , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Allosteric Regulation , Ammonia/metabolism , Glutaminase/antagonists & inhibitors , Glutamine/metabolism , Hydrolysis , Kinetics , Uridine Triphosphate/metabolism , Uridine Triphosphate/pharmacology
15.
Biochim Biophys Acta ; 518(2): 267-76, 1978 Apr 27.
Article in English | MEDLINE | ID: mdl-350279

ABSTRACT

5a,6-Anhydrotetracycline was discovered to be unique among several tetracycline derivatives tested in its ability to inhibit RNA accumulation in vivo at low concentration (20 microgram/ml and less). In addition, in vivo protein, DNA, and guanosine 5'-diphosphate 3'-diphosphate (ppGpp) synthesis were completely inhibited by 20 microgram/ml 5a,6-anhydrotetracycline. ppGpp decay in a spoT strain was inhibited by 20 microgram/ml 5a,6-anhydrotef RNA synthesis by a 5a,6-anhydrotetracycline may be due, in part, to reduced UTP and CTP synthesis. The effects of tetracyclines on in vitro ppGpp synthesis by crude stringent factor in the absence of ribosomes were investigated. It was determined that of six tetracyclines tested, four strongly inhibited the reaction (oxytetracycline, chlorotetracycline, dedimethylaminotetracycline, and tetracycline) whereas 5a,6-anhydrotetracycline gave a moderate inhibition and alpha-6-deoxyoxytetracycline resulted in only a slight reduction in ppGpp synthesis. It is proposed that tetracyclines interfere with factors involved in ppGpp metabolism and function.


Subject(s)
Escherichia coli/metabolism , Guanine Nucleotides/metabolism , Guanosine Tetraphosphate/metabolism , RNA, Bacterial/biosynthesis , Tetracyclines/pharmacology , Bacterial Proteins/biosynthesis , Cytidine Triphosphate/biosynthesis , DNA, Bacterial/biosynthesis , Escherichia coli/drug effects , Peptide Biosynthesis , Phenylalanine/metabolism , Uridine Triphosphate/biosynthesis
16.
Biochim Biophys Acta ; 755(2): 217-24, 1983 Jan 25.
Article in English | MEDLINE | ID: mdl-6403046

ABSTRACT

Nucleotide biosynthesis in Novikoff hepatoma cells is markedly altered by a variety of chemical mutagens, whether the mechanism of mutagenesis is by base substitution, covalent binding (adduct formation), intercalation, or cross-linking of DNA. The compounds investigated (N-methyl-N'-nitro-N-nitrosoguanidine, 4-nitroquinoline 1-oxide, 9-aminoacridine, and mitomycin C), at concentrations that cause some inhibition of RNA and DNA synthesis, bring about a large increase in the pool levels of all four nucleoside triphosphates. At the same time, reactions leading to the synthesis of CTP from exogenous uridine and GTP and ATP from exogenous hypoxanthine are severely inhibited. The formation of UTP from uridine and ATP from adenosine, by more direct phosphorylation reactions, appears relatively unaffected. The increase in nucleotide pool size cannot be accounted for by a corresponding increase in de novo purine and pyrimidine nucleotide synthesis, as experiments with labeled formate and aspartate show similar inhibitions by the mutagens. With the salvage precursors, [3H]uridine and [3H]hypoxanthine, the mutagens can produce a widely divergent reduction in the labeling of RNA-CMP versus RNA-UMP and of RNA-GMP versus RNA-AMP, mostly a result of these agents causing large differences in the specific activities of the respective triphosphate precursors. These observations suggest that, in addition to the reactions with DNA, nucleotide biosynthesis could be another important biochemical target of chemical mutagens.


Subject(s)
Adenosine Triphosphate/biosynthesis , Cytidine Triphosphate/biosynthesis , Cytosine Nucleotides/biosynthesis , Guanosine Triphosphate/biosynthesis , Liver Neoplasms, Experimental/metabolism , Mutagens/pharmacology , Uracil Nucleotides/biosynthesis , Uridine Triphosphate/biosynthesis , 4-Nitroquinoline-1-oxide/pharmacology , Aminacrine/pharmacology , Animals , DNA Replication/drug effects , Kinetics , Methylnitronitrosoguanidine/pharmacology , Mitomycin , Mitomycins/pharmacology , Rats , Transcription, Genetic/drug effects
17.
Leukemia ; 8(8): 1375-8, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8057676

ABSTRACT

The pattern of incorporation of [14C]uridine showed that in MOLT-3 cells an increased proportion of CTP was synthesized via CTP synthetase, compared to proliferating normal human T lymphocytes at a physiological concentration of cytidine (< 0.5 microM). Furthermore, in the proliferating normal human T lymphocytes similar patterns of incorporation of [14C]uridine were observed in the presence of the physiological concentration of cytidine and after addition of 2 microM of cytidine. In contrast, in the MOLT-3 cells after addition of 2 microM of cytidine the proportion of CTP synthesized by conversion of UTP into CTP was substantially decreased, whereas the salvage of cytidine was proportionally increased. We conclude that the reutilization of uridine is a preferred route in the synthesis of CTP for MOLT-3 cells at physiological concentrations of uridine and cytidine, whereas in proliferating normal human T lymphocytes CTP is largely synthesized through reutilization of cytidine. This difference in salvage of pyrimidine ribonucleosides may be exploited for selective chemotherapy.


Subject(s)
Carbon-Nitrogen Ligases , Cytidine Triphosphate/biosynthesis , Ligases/metabolism , T-Lymphocytes/enzymology , Uridine Kinase/metabolism , Carbon Radioisotopes , Cell Line , Humans , Models, Biological , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Tumor Cells, Cultured , Uridine/metabolism
18.
FEBS Lett ; 527(1-3): 229-33, 2002 Sep 11.
Article in English | MEDLINE | ID: mdl-12220665

ABSTRACT

In this paper, it is demonstrated that all-trans, 9-cis and 13-cis retinoic acid (RA) decreased the sensitivity of SK-N-BE(2)c neuroblastoma cells towards the chemotherapeutic agent cyclopentenyl cytosine (CPEC), a potent inhibitor of cytosine-5'-triphosphate synthetase. Retinoic acid attenuated CPEC-induced apoptosis as reflected by a decreased caspase-3 induction. Retinoic acid decreased the accumulation of CPEC, whereas the salvage of cytidine was strongly increased. Metabolic labeling studies using [(3)H]uridine showed a strongly decreased biosynthesis of CTP via CTP synthetase. Retinoic acid likely confers resistance of neuroblastoma cells to CPEC in part by slowing down proliferation, and in part by shifting the synthesis of CTP towards the salvage of cytidine, thereby bypassing CTP synthetase.


Subject(s)
Antineoplastic Agents/pharmacology , Cytidine/analogs & derivatives , Cytidine/pharmacology , Neuroblastoma/drug therapy , Tretinoin/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cytidine Triphosphate/biosynthesis , Drug Interactions , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Polyphosphates/metabolism , Tumor Cells, Cultured
19.
Eur J Cancer ; 31A(1): 108-12, 1995.
Article in English | MEDLINE | ID: mdl-7695960

ABSTRACT

The role of cytidine triphosphate (CTP) synthetase (EC 6.3.4.2.) in the pyrimidine ribonucleotide metabolism of MOLT-3 human T-ALL cell line cells and normal human T lymphocytes during the cell cycle traverse was studied. Highly pure G1-phase samples and samples enriched in S-phase cells were obtained by counterflow centrifugation. The activity of CTP synthetase in situ, measured in pulse-chase experiments, was similar in the G1-phase and S-phase MOLT-3 cells. In contrast, in S-phase T lymphocytes, an increased activity of CTP synthetase was observed compared with G1-phase T lymphocytes. Nevertheless, the MOLT-3 samples showed an increased activity of CTP synthetase in comparison with either G1-phase or S-phase enriched samples of normal T lymphocytes. Therefore, the increased activity of CTP synthetase of MOLT-3 cells is a cell cycle-independent feature, whereas among normal T lymphocytes, the increase in activity of CTP synthetase that arises after a growth stimulus is more prominent in the S-phase.


Subject(s)
Carbon-Nitrogen Ligases , Leukemia-Lymphoma, Adult T-Cell/enzymology , Ligases/metabolism , T-Lymphocytes/enzymology , Cell Cycle , Cytidine Triphosphate/biosynthesis , Humans , Nucleotides/metabolism , Tumor Cells, Cultured/enzymology , Uridine/metabolism
20.
Biochem Pharmacol ; 34(14): 2535-9, 1985 Jul 15.
Article in English | MEDLINE | ID: mdl-4015694

ABSTRACT

The mechanism of action of the cyclopentenyl analogue of cytidine, cCyd, was investigated in human colon carcinoma cell line HT-29. Upon exposure of cells to 10(-6)M cCyd, cell viability was reduced to 20% of control, whereas cytocidal activity was not present after 2 hr of drug exposure. Cell lethality was partially reversible by Urd, Cyd or dCyd at 10(-6)M cCyd, and fully reversible by these nucleosides at 2.5 X 10(-7)M cCyd. The incorporation of [14C]dThd and [3H]Urd into DNA and RNA was inhibited by 50% by exposure for 2 hr to 2.5 X 10(-7) and 1.5 X 10(-6)M cCyd respectively. Upon 24 hr of drug exposure, the IC50 for RNA synthesis was reduced 2.5-fold, whereas DNA synthesis was almost totally inhibited. cCyd produced a rapid and preferential reduction of CTP synthesis with a half-life of 1 hr at 10(-6)M drug. The IC50 of cCyd for reducing CTP concentrations after 2 hr of drug exposure was 4 X 10(-7)M. Concomitant with the reduction of CTP levels was the inhibition of transcription of rRNA and, to a lesser extent, tRNA, without changes in the processing nucleolar RNA. No changes in the size of DNA were produced following treatment with cCyd. These results indicate that cCyd is a potent and rapid inhibitor of CTP synthesis and that this effect correlates with its cytocidal activity.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Cytidine Triphosphate/biosynthesis , Cytidine/analogs & derivatives , Cytosine Nucleotides/biosynthesis , Cell Line , Cell Survival/drug effects , Cytidine/pharmacology , DNA, Neoplasm/biosynthesis , Humans , RNA, Neoplasm/biosynthesis , Uridine/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL