Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Elife ; 3: e03638, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-25030911

ABSTRACT

CTP Synthetase (CtpS) is a universally conserved and essential metabolic enzyme. While many enzymes form small oligomers, CtpS forms large-scale filamentous structures of unknown function in prokaryotes and eukaryotes. By simultaneously monitoring CtpS polymerization and enzymatic activity, we show that polymerization inhibits activity, and CtpS's product, CTP, induces assembly. To understand how assembly inhibits activity, we used electron microscopy to define the structure of CtpS polymers. This structure suggests that polymerization sterically hinders a conformational change necessary for CtpS activity. Structure-guided mutagenesis and mathematical modeling further indicate that coupling activity to polymerization promotes cooperative catalytic regulation. This previously uncharacterized regulatory mechanism is important for cellular function since a mutant that disrupts CtpS polymerization disrupts E. coli growth and metabolic regulation without reducing CTP levels. We propose that regulation by large-scale polymerization enables ultrasensitive control of enzymatic activity while storing an enzyme subpopulation in a conformationally restricted form that is readily activatable.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Cytidine Triphosphate/biosynthesis , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Recombinant Fusion Proteins/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Expression , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
7.
Dis Model Mech ; 6(2): 521-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22996645

ABSTRACT

Anti-cancer drug development involves enormous expenditure and risk. For rapid and economical identification of novel, bioavailable anti-tumour chemicals, the use of appropriate in vivo tumour models suitable for large-scale screening is key. Using a Drosophila Ras-driven tumour model, we demonstrate that tumour overgrowth can be curtailed by feeding larvae with chemicals that have the in vivo pharmacokinetics essential for drug development and known efficacy against human tumour cells. We then develop an in vivo 96-well plate chemical screening platform to carry out large-scale chemical screening with the tumour model. In a proof-of-principle pilot screen of 2000 compounds, we identify the glutamine analogue, acivicin, a chemical with known activity against human tumour cells, as a potent and specific inhibitor of Drosophila tumour formation. RNAi-mediated knockdown of candidate acivicin target genes implicates an enzyme involved in pyrimidine biosynthesis, CTP synthase, as a possible crucial target of acivicin-mediated inhibition. Thus, the pilot screen has revealed that Drosophila tumours are glutamine-dependent, which is an emerging feature of many human cancers, and has validated the platform as a powerful and economical tool for in vivo chemical screening. The platform can also be adapted for use with other disease models, thus offering widespread applications in drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drosophila melanogaster/drug effects , Drug Screening Assays, Antitumor/methods , Neoplasms/drug therapy , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Biological Availability , Cell Proliferation/drug effects , Cytidine Triphosphate/biosynthesis , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drosophila melanogaster/cytology , Glutamine/metabolism , Isoxazoles/pharmacology , Isoxazoles/therapeutic use , Pharmacogenetics , Pilot Projects
9.
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
10.
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
11.
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
12.
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
13.
Cancer Chemother Pharmacol ; 63(1): 55-64, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18305939

ABSTRACT

AIM: This study aims to determine if intracellular formation of gemcitabine triphosphate (dFdCTP), an active metabolite of gemcitabine, is saturable at doses used for treatment of Asian patients with lung cancer. METHODS: From a phase II trial, plasma concentrations of gemcitabine, its inactive metabolite 2'-2'-difluorodeoxyuridine (dFdU), and mononuclear cell concentrations of gemcitabine-triphosphate were measured in 56 and 33 patients, respectively. The pharmacokinetics of gemcitabine and metabolites were modeled using nonlinear mixed effects modeling (NONMEM). A reduced dataset of ten randomly selected patients was employed to compare first-order and saturable formation of dFdCTP from gemcitabine. RESULTS: The median population clearance estimate for dFdCTP formation with the full dataset was 70.2 L/h/70 kg/1.7 m. Modeling Michaelis-Menten formation of dFdCTP on a reduced dataset estimated K(m) to be 3.6 times higher than the maximum gemcitabine concentration (72.2 microM) measured in this study. CONCLUSIONS: The results showed that first-order and nonsaturable clearance described intracellular dFdCTP formation at clinically applied doses of gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cytidine Triphosphate/analogs & derivatives , Deoxycytidine/analogs & derivatives , Lung Neoplasms/metabolism , Prodrugs/pharmacokinetics , Adipose Tissue , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biotransformation , Body Weight , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Clinical Trials, Phase II as Topic , Creatinine/blood , Cytidine Triphosphate/biosynthesis , Cytidine Triphosphate/pharmacology , Deoxycytidine/pharmacokinetics , Female , Humans , Lung Neoplasms/drug therapy , Male , Middle Aged , Models, Biological , Prospective Studies , Randomized Controlled Trials as Topic , Gemcitabine
14.
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
15.
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
16.
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
17.
Arch Biochem Biophys ; 424(1): 105-11, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15019842

ABSTRACT

CTP synthase catalyses the reaction: glutamine+UTP+ATP --> glutamate+CTP+ADP+P(i). The reaction is greatly stimulated by the allosteric binding of GTP. In addition to glutamine that is hydrolysed by the enzyme to ammonia and glutamate, CTP synthase will also utilise external sources of amino donors such as NH(4)Cl. This reaction is no longer dependent on allosteric activation by GTP. Hydroxylamine is also a substrate for Lactococcus lactis CTP synthase and results in the formation of N4-OH CTP. This product has the feature that it absorbs at 300nm where CTP absorption was shown to be greatly reduced and enabled the determination of N4-OH CTP formation in the presence of CTP synthesis derived from glutamine hydrolysis. Differences in initial rates determined for the hydroxylamine dependent reaction at 291nm in the presence and absence of glutamine and GTP were ascribed to simultaneous CTP and N4-OH CTP synthesis in the presence of these compounds. A characterisation of the apparent inhibition by GTP and glutamine of N4-OH CTP synthesis determined at 300nm showed that glutamine dependent CTP synthesis occurs at a rate of about 60% of that in the absence of hydroxylamine. GTP dependent inhibition of the ammonium chloride dependent reaction of L. lactis CTP synthase by the glutamine analog glutamate gamma-semialdehyde showed a partial inhibition with a maximum inhibition of about 60%. These results are interpreted in terms of a "half of the sites" mechanism for glutamine hydrolysis on CTP synthase.


Subject(s)
Ammonia/metabolism , Carbon-Nitrogen Ligases/metabolism , Glutamine/metabolism , Hydroxylamine/metabolism , Lactococcus lactis/enzymology , Uridine Triphosphate/metabolism , Binding, Competitive , Carbon-Nitrogen Ligases/antagonists & inhibitors , Catalysis , Cytidine Triphosphate/analogs & derivatives , Cytidine Triphosphate/biosynthesis , Enzyme Inhibitors/pharmacology , Glutamates/pharmacology , Guanosine Triphosphate/metabolism , Hydrolysis , Kinetics , Lactococcus lactis/metabolism , Protein Binding , Protein Subunits/metabolism , Solutions , Uridine Triphosphate/analogs & derivatives
18.
J Biol Chem ; 278(23): 20785-94, 2003 Jun 06.
Article in English | MEDLINE | ID: mdl-12670958

ABSTRACT

The Saccharomyces cerevisiae URA7-encoded CTP synthetase is phosphorylated and stimulated by protein kinase C. We examined the hypothesis that Ser36, Ser330, Ser354, and Ser454, contained in a protein kinase C sequence motif in CTP synthetase, were target sites for the kinase. Synthetic peptides containing a phosphorylation motif at these serine residues served as substrates for protein kinase C in vitro. Ser --> Ala (S36A, S330A, S354A, and S454A) mutations in CTP synthetase were constructed by site-directed mutagenesis and expressed normally in a ura7 ura8 double mutant that lacks CTP synthetase activity. The CTP synthetase activity in extracts from cells bearing the S36A, S354A, and S454A mutant enzymes was reduced when compared with cells bearing the wild type enzyme. Kinetic analysis of purified mutant enzymes showed that the S36A and S354A mutations caused a decrease in the Vmax of the reaction. This regulation could be attributed in part by the effects phosphorylation has on the nucleotide-dependent oligomerization of CTP synthetase. In contrast, CTP synthetase activity in cells bearing the S330A mutant enzyme was elevated, and kinetic analysis of purified enzyme showed that the S330A mutation caused an elevation in the Vmax of the reaction. In vitro data indicated that phosphorylation of CTP synthetase at Ser330 affected the phosphorylation of the enzyme at another site. The phosphorylation of CTP synthetase at Ser36, Ser330, Ser354, and Ser454 residues was physiologically relevant. Cells bearing the S36A, S354A, and S454A mutations had reduced CTP levels, whereas cells with the S330A mutation had elevated CTP levels. The alterations in CTP levels correlated with the regulatory effects CTP has on the pathways responsible for the synthesis of the membrane phospholipid phosphatidylcholine.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Cytidine Triphosphate/metabolism , Phosphatidylcholines/metabolism , Saccharomyces cerevisiae/enzymology , Carbon-Nitrogen Ligases/genetics , Cytidine Triphosphate/biosynthesis , Enzyme Activation/genetics , Mutagenesis, Site-Directed , Phosphatidylcholines/biosynthesis , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C/metabolism , Saccharomyces cerevisiae/genetics , Serine/metabolism , Substrate Specificity
19.
Biochem J ; 369(Pt 3): 497-507, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12383057

ABSTRACT

CTP synthase catalyses the ATP-dependent formation of CTP from UTP using either NH(3) or L-glutamine as the nitrogen source. GTP is required as an allosteric effector to promote glutamine hydrolysis. In an attempt to identify nucleotide-binding sites, scanning alanine mutagenesis was conducted on a highly conserved region of amino acid sequence (residues 102-118) within the synthase domain of Escherichia coli CTP synthase. Mutant K102A CTP synthase exhibited wild-type activity with respect to NH(3) and glutamine; however, the R105A, D107A, L109A and G110A enzymes exhibited wild-type NH(3)-dependent activity and affinity for glutamine, but impaired glutamine-dependent CTP formation. The E103A, R104A and H118A enzymes exhibited no glutamine-dependent activity and were only partially active with NH(3). Although these observations were compatible with impaired activation by GTP, the apparent affinity of the D107A, L109A and G110A enzymes for GTP was reduced only 2-4-fold, suggesting that these residues do not play a significant role in GTP binding. In the presence of GTP, the k (cat) values for glutamine hydrolysis by the D107A and L109A enzymes were identical with that of wild-type CTP synthase. Overall, the kinetic properties of L109A CTP synthase were consistent with an uncoupling of glutamine hydrolysis from CTP formation that occurs because an NH(3) tunnel has its normal structure altered or fails to form. L109F CTP synthase was prepared to block totally the putative NH(3) tunnel; however, this enzyme's rate of glutamine-dependent CTP formation and glutaminase activity were both impaired. In addition, we observed that mutation of amino acids located between residues 102 and 118 in the synthase domain can affect the enzyme's glutaminase activity, suggesting that these residues interact with residues in the glutamine amide transfer domain because they are in close proximity or via a conformationally dependent signalling mechanism.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Cytidine Triphosphate/biosynthesis , Escherichia coli Proteins/metabolism , Glutamine/metabolism , Alanine/genetics , Amino Acid Sequence , Aspartic Acid/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/genetics , Circular Dichroism , Conserved Sequence , Dimerization , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Guanosine Triphosphate/metabolism , Hydrolysis , Leucine/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed
20.
Eur J Biochem ; 269(19): 4772-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12354108

ABSTRACT

CTP synthase catalyzes the reaction glutamine + UTP + ATP --> glutamate + CTP + ADP + Pi. The rate of the reaction is greatly enhanced by the allosteric activator GTP. We have studied the glutaminase half-reaction of CTP synthase from Lactococcus lactis and its response to the allosteric activator GTP and nucleotides that bind to the active site. In contrast to what has been found for the Escherichia coli enzyme, GTP activation of the L. lactis enzyme did not result in similar kcat values for the glutaminase activity and glutamine hydrolysis coupled to CTP synthesis. GTP activation of the glutaminase reaction never reached the levels of GTP-activated CTP synthesis, not even when the active site was saturated with UTP and the nonhydrolyzeable ATP-binding analog adenosine 5'-[gamma-thio]triphosphate. Furthermore, under conditions where the rate of glutamine hydrolysis exceeded that of CTP synthesis, GTP would stimulate CTP synthesis. These results indicate that the L. lactis enzyme differs significantly from the E. coli enzyme. For the E. coli enzyme, activation by GTP was found to stimulate glutamine hydrolysis and CTP synthesis to the same extent, suggesting that the major function of GTP binding is to activate the chemical steps of glutamine hydrolysis. An alternative mechanism for the action of GTP on L. lactis CTP synthase is suggested. Here the binding of GTP to the allosteric site promotes coordination of the phosphorylation of UTP and hydrolysis of glutamine for optimal efficiency in CTP synthesis rather than just acting to increase the rate of glutamine hydrolysis itself.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Glutaminase/metabolism , Lactococcus lactis/enzymology , Allosteric Regulation , Calorimetry , Carbon-Nitrogen Ligases/antagonists & inhibitors , Carbon-Nitrogen Ligases/chemistry , Cytidine Triphosphate/biosynthesis , Glutamine/metabolism , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Hydrolysis , Kinetics , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...