Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Protein Sci ; 29(11): 2189-2200, 2020 11.
Article in English | MEDLINE | ID: mdl-32864839

ABSTRACT

Metabolic pathways in cancer cells typically become reprogrammed to support unconstrained proliferation. These abnormal metabolic states are often accompanied by accumulation of high concentrations of ATP in the cytosol, a phenomenon known as the Warburg Effect. However, how high concentrations of ATP relate to the functional state of proteins is poorly understood. Here, we comprehensively studied the influence of ATP levels on the functional state of the human enzyme, uridine phosphorylase I (hUP1), which is responsible for activating the chemotherapeutic pro-drug, 5-fluorouracil. We found that elevated levels of ATP decrease the stability of hUP1, leading to the loss of its proper folding and function. We further showed that the concentration of hUP1 exerts a critical influence on this ATP-induced destabilizing effect. In addition, we found that ATP interacts with hUP1 through a partially unfolded state and accelerates the rate of hUP1 unfolding. Interestingly, some structurally similar metabolites showed similar destabilization effects on hUP1. Our findings suggest that metabolites can alter the folding and function of a human protein, hUP1, through protein destabilization. This phenomenon may be relevant in studying the functions of proteins that exist in the specific metabolic environment of a cancer cell.


Subject(s)
Adenosine Triphosphate/chemistry , Fluorouracil/chemistry , Protein Unfolding , Uridine Phosphorylase/chemistry , Adenosine Triphosphate/metabolism , Enzyme Stability , Humans , Uridine Phosphorylase/metabolism
2.
Sci Rep ; 10(1): 9051, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32493959

ABSTRACT

Uridine phosphorylase (UP) is a key enzyme of pyrimidine salvage pathways that enables the recycling of endogenous or exogenous-supplied pyrimidines and plays an important intracellular metabolic role. Here, we biochemically and structurally characterized two evolutionarily divergent uridine phosphorylases, PcUP1 and PcUP2 from the oomycete pathogen Phytophthora capsici. Our analysis of other oomycete genomes revealed that both uridine phosphorylases are present in Phytophthora and Pythium genomes, but only UP2 is seen in Saprolegnia spp. which are basal members of the oomycetes. Moreover, uridine phosphorylases are not found in obligate oomycete pathogens such as Hyaloperonospora arabidopsidis and Albugo spp. PcUP1 and PcUP2 are upregulated 300 and 500 fold respectively, within 90 min after infection of pepper leaves. The crystal structures of PcUP1 in ligand-free and in complex with uracil/ribose-1-phosphate, 2'-deoxyuridine/phosphate and thymidine/phosphate were analyzed. Crystal structure of this uridine phosphorylase showed strict conservation of key residues in the binding pocket. Structure analysis of PcUP1 with bound ligands, and site-directed mutagenesis of key residues provide additional support for the "push-pull" model of catalysis. Our study highlights the importance of pyrimidine salvage during the earliest stages of infection.


Subject(s)
Phytophthora/metabolism , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/metabolism , Binding Sites/physiology , Catalysis , Catalytic Domain/physiology , Crystallography, X-Ray/methods , Deoxyuridine/chemistry , Deoxyuridine/metabolism , Ligands , Pyrimidines/chemistry , Pyrimidines/metabolism , Ribosemonophosphates/chemistry , Ribosemonophosphates/metabolism , Thymidine/chemistry , Thymidine/metabolism , Uracil/chemistry , Uracil/metabolism , Uridine/chemistry , Uridine/metabolism
3.
Arch Biochem Biophys ; 634: 11-20, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28917600

ABSTRACT

Uridine phosphorylase is one of the critical enzymes in the pyrimidine salvage pathway. Cells regenerate uridine for nucleotide metabolism by incorporating uracil with ribose-1-phosphate with this enzyme. Recent studies indicate that Escherichia coli uridine phosphorylase is destabilized in the presence of ATP. However, the mechanism underlying the destabilization process and its influence on uridine phosphorylase function remain to be established. Here, we comprehensively investigated the effects of ATP on protein folding and function of Escherichia coli uridine phosphorylase. Our results demonstrate that ATP apparently decreases the stability of uridine phosphorylase in a concentration-dependent manner. Additionally, simply increasing the level of ATP led to a reduction of enzymatic activity to complete inhibition. Further studies showed that uridine phosphorylase accumulates as a partially unfolded state in the presence of ATP. Moreover, ATP specifically accelerated the unfolding rate of uridine phosphorylase with no observable effects on the refolding process. Our preliminary findings suggest that ATP can alter the protein folding and function of enzymes via apparent destabilization. This mechanism may be significant for proteins functioning under conditions of high levels of ATP, such as cancer cell environments.


Subject(s)
Adenosine Triphosphate/chemistry , Escherichia coli/enzymology , Models, Chemical , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/ultrastructure , Computer Simulation , Enzyme Activation , Enzyme Stability , Protein Conformation , Protein Folding
4.
Nucleosides Nucleotides Nucleic Acids ; 36(2): 107-121, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27846376

ABSTRACT

Twenty five uridine analogues have been tested and compared with uridine with respect to their potency to bind to E. coli uridine phosphorylase. The kinetic constants of the phosphorolysis reaction of uridine derivatives modified at 2'-, 3'- and 5'-positions of the sugar moiety and 2-, 4-, 5- and 6-positions of the heterocyclic base were determined. The absence of the 2'- or 5'-hydroxyl group is not crucial for the successful binding and phosphorolysis. On the other hand, the absence of both the 2'- and 5'-hydroxyl groups leads to the loss of substrate binding to the enzyme. The same effect was observed when the 3'-hydroxyl group is absent, thus underlining the key role of this group. Our data shed some light on the mechanism of ribo- and 2'-deoxyribonucleoside discrimination by E. coli uridine phosphorylase and E. coli thymidine phosphorylase. A comparison of the kinetic results obtained in the present study with the available X-ray structures and analysis of hydrogen bonding in the enzyme-substrate complex demonstrates that uridine adopts an unusual high-syn conformation in the active site of uridine phosphorylase.


Subject(s)
Escherichia coli Proteins/metabolism , Uridine Phosphorylase/metabolism , Uridine/chemistry , Uridine/metabolism , Catalytic Domain , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Hydrogen Bonding , Kinetics , Models, Molecular , Protein Conformation , Substrate Specificity , Uridine Phosphorylase/chemistry
5.
Biochimie ; 125: 12-22, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26898674

ABSTRACT

Reports of Schistosoma mansoni strains resistant to praziquantel, the only therapeutic strategy available for the treatment of schistosomiasis, have motivated the scientific community towards the search for new possible therapies. Biochemical characterization of the parasite's metabolism is an essential component for the rational development of new therapeutic alternatives. One of the so far uncharacterized enzymes is uridine phosphorylase (UP) (EC 2.4.2.3), for which the parasite genome presents two isoforms (SmUPa and SmUPb) that share 92% sequence identity. In this paper, we present crystal structures for SmUPa and SmUPb in their free states as well as bound to different ligands. This we have complemented by enzyme kinetic characterization and phylogenetic analyses. Both enzymes present an overall fold and active site structure similar to other known UPs. The kinetic analyses showed conclusively that SmUPa is a regular uridine phosphorylase but by contrast SmUPb presented no detectable activity. This is particularly noteworthy given the high level of sequence identity between the two isoforms and is probably the result of the significant differences observed for SmUPb in the vicinity of the active site itself, suggesting that it is not a UP at all. On the other hand, it was not possible to identify an alternative function for SmUPb, although our phylogenetic analyses and expression data suggest that SmUPb is still functional and plays a role in parasite metabolism. The unusual UPb isoform may open up new opportunities for understanding unique features of S. mansoni metabolism.


Subject(s)
Helminth Proteins/chemistry , Schistosoma mansoni/enzymology , Uridine Phosphorylase/chemistry , Animals , Crystallography, X-Ray , Isoenzymes , Protein Domains
6.
Acta Crystallogr D Struct Biol ; 72(Pt 2): 203-10, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26894668

ABSTRACT

Uridine phosphorylase (UP; EC 2.4.2.3), a key enzyme in the pyrimidine-salvage pathway, catalyzes the reversible phosphorolysis of uridine to uracil and ribose 1-phosphate. The structure of the C212S mutant of uridine phosphorylase from the facultatively aerobic Gram-negative γ-proteobacterium Shewanella oneidensis MR-1 (SoUP) was determined at 1.68 Šresolution. A comparison of the structures of the mutant and the wild-type enzyme showed that one dimer in the mutant hexamer differs from all other dimers in the mutant and wild-type SoUP (both in the free form and in complex with uridine). The key difference is the `maximum open' state of one of the subunits comprising this dimer, which has not been observed previously for uridine phosphorylases. Some conformational features of the SoUP dimer that provide access of the substrate into the active site are revealed. The binding of the substrate was shown to require the concerted action of two subunits of the dimer. The changes in the three-dimensional structure induced by the C212S mutation account for the lower affinity of the mutant for inorganic phosphate, while the affinity for uridine remains unchanged.


Subject(s)
Bacterial Proteins/chemistry , Shewanella/enzymology , Uridine Phosphorylase/chemistry , Catalytic Domain , Crystallography, X-Ray , Hydrogen Bonding , Kinetics , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Substrate Specificity , Uridine/chemistry
7.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 12): 3310-9, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25478848

ABSTRACT

Uridine phosphorylase (UP; EC 2.4.2.3), a key enzyme in the pyrimidine-salvage pathway, catalyzes the reversible phosphorolysis of uridine to uracil and ribose 1-phosphate. Expression of UP from Shewanella oneidensis MR-1 (SoUP) was performed in Escherichia coli. The high-resolution X-ray structure of SoUP was solved in the free form and in complex with uridine. A crystal of SoUP in the free form was grown under microgravity and diffracted to ultrahigh resolution. Both forms of SoUP contained sulfate instead of phosphate in the active site owing to the presence of ammonium sulfate in the crystallization solution. The latter can be considered as a good mimic of phosphate. In the complex, uridine adopts a high-syn conformation with a nearly planar ribose ring and is present only in one subunit of the hexamer. A comparison of the structures of SoUP in the free form and in complex with the natural substrate uridine showed that the subunits of the hexamer are not identical, with the active sites having either an open or a closed conformation. In the monomers with the closed conformation, the active sites in which uridine is absent contain a glycerol molecule mimicking the ribose moiety of uridine.


Subject(s)
Shewanella/enzymology , Uridine Phosphorylase/chemistry , Uridine/metabolism , Catalytic Domain , Crystallography, X-Ray , Gram-Negative Bacterial Infections/microbiology , Humans , Protein Conformation , Shewanella/chemistry , Shewanella/metabolism , Uridine/chemistry , Uridine Phosphorylase/metabolism
8.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 1): 60-3, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24419619

ABSTRACT

Uridine phosphorylase catalyzes the phosphorolysis of ribonucleosides, with the nitrogenous base and ribose 1-phosphate as products. Additionally, it catalyzes the reverse reaction of the synthesis of ribonucleosides from ribose 1-phosphate and a nitrogenous base. However, the enzyme does not catalyze the synthesis of nucleosides when the substrate is a nitrogenous base substituted at the 6-position, such as 6-methyluracil (6-MU). In order to explain this fact, it is essential to investigate the three-dimensional structure of the complex of 6-MU with uridine phosphorylase. 6-MU is a pharmaceutical agent that improves tissue nutrition and enhances cell regeneration by normalization of nucleotide exchange in humans. 6-MU is used for the treatment of diseases of the gastrointestinal tract, including infectious diseases. Here, procedures to obtain the uridine phosphorylase from the pathogenic bacterium Vibrio cholerae (VchUPh), purification of this enzyme, crystallization of the complex of VchUPh with 6-MU, and X-ray data collection and preliminary X-ray analysis of the VchUPh-6-MU complex at atomic resolution are reported.


Subject(s)
Uracil/analogs & derivatives , Uridine Phosphorylase/chemistry , Vibrio cholerae/enzymology , Binding Sites , Biocatalysis , Crystallization , Crystallography, X-Ray , Models, Molecular , Uracil/chemistry
10.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 68(Pt 11): 1387-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23143255

ABSTRACT

Uridine phosphorylase (UDP, EC 2.4.2.3), a key enzyme in the pyrimidine salvage pathway, catalyses the reversible phosphorolysis of uridine to uracil and ribose 1-phosphate. The gene expression of UDP from Shewanella oneidensis MR-1 was performed in the recipient strain Escherichia coli. The UDP protein was crystallized on earth (in the free form and in complex with uridine as the substrate) by the hanging-drop vapour-diffusion method at 296 K and under microgravity conditions (in the free form) aboard the Russian Segment of the International Space Station by the capillary counter-diffusion method. The data sets were collected to a resolution of 1.9 Šfrom crystals of the free form grown on earth, 1.6 Šfrom crystals of the complex with uridine and 0.95 Šfrom crystals of the free form grown under microgravity. All crystals belong to the space group P2(1) and have similar unit-cell parameters. The crystal of uridine phosphorylase grown under microgravity diffracted to ultra-high resolution and gave high-quality X-ray diffraction data.


Subject(s)
Bacterial Proteins/chemistry , Shewanella/enzymology , Uridine Phosphorylase/chemistry , Crystallization , Crystallography, X-Ray , Uridine/chemistry , Weightlessness
11.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 68(Pt 11): 1394-7, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23143257

ABSTRACT

A high-resolution structure of the complex of Vibrio cholerae uridine phosphorylase (VchUPh) with its physiological ligand thymidine is important in order to determine the mechanism of the substrate specificity of the enzyme and for the rational design of pharmacological modulators. Here, the expression and purification of VchUPh and the crystallization of its complex with thymidine are reported. Conditions for crystallization were determined with an automated Cartesian Dispensing System using The Classics, MbClass and MbClass II Suites crystallization kits. Crystals of the VchUPh-thymidine complex (of dimensions ∼200-350 µm) were grown by the sitting-drop vapour-diffusion method in ∼7 d at 291 K. The crystallization solution consisted of 1.5 µl VchUPh (15 mg ml(-1)), 1 µl 0.1 M thymidine and 1.5 µl reservoir solution [15%(w/v) PEG 4000, 0.2 M MgCl(2).6H2O in 0.1 M Tris-HCl pH 8.5]. The crystals diffracted to 2.12 Šresolution and belonged to space group P2(1) (No. 4), with unit-cell parameters a=91.80, b=95.91, c=91.89 Å, ß=119.96°. The Matthews coefficient was calculated as 2.18 Å3 Da(-1); the corresponding solvent content was 43.74%.


Subject(s)
Bacterial Proteins/chemistry , Thymidine/chemistry , Uridine Phosphorylase/chemistry , Vibrio cholerae/enzymology , Amino Acid Motifs , Bacterial Proteins/isolation & purification , Catalytic Domain , Crystallization , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Models, Molecular , Uridine Phosphorylase/isolation & purification
12.
Gene ; 510(2): 154-61, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22967797

ABSTRACT

Nucleoside phosphorylases are essential for the salvage and catabolism of nucleotides in bacteria and other organisms, and members of this enzyme superfamily have been of interest for the development of antimicrobial and cancer therapies. The nucleotide phosphorylase superfamily 1 encompasses a number of different enzymes which share a general superfold and catalytic mechanism, while they differ in the nature of the nucleophiles used and in the nature of characteristic active site residues. Recently, one subfamily, the uridine phosphorylases, has been subdivided into two types which differ with respect to the mechanism of transition state stabilization, as dictated by differences in critical amino acid residues. Little is known about the phylogenetic distribution and relationship of the two different types, as well as the relationship to other NP-1 superfamily members. Here comparative genomic analysis illustrates that UP-1s and UP-2s fall into monophyletic groups and are biased with respect to species representation. UP-1 evolved in Gram negative bacteria, while Gram positive species tend to predominantly contain UP-2. PNP (a sister clade to all UPs) contains both Gram positive and Gram negative species. The findings imply that the nucleoside phosphorylase superfamily 1 evolved through a series of three important duplications, leading to the separate, monophyletic enzyme families, coupled to individual lateral transfer events. Extensive horizontal transfer explains the occurrence of unexpected uridine phosphorylases in some genomes. This study provides a basis for understanding the evolution of uridine and purine nucleoside phosphorylases with respect to DNA/RNA metabolism and with potential utility in the design of antimicrobial and anti-tumor drugs.


Subject(s)
Bacteria/enzymology , Bacteria/genetics , Evolution, Molecular , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Catalytic Domain , Escherichia coli/enzymology , Escherichia coli/genetics , Pentosyltransferases/chemistry , Pentosyltransferases/classification , Phylogeny , Purine-Nucleoside Phosphorylase/chemistry , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/genetics , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/classification , Uridine Phosphorylase/genetics , Uridine Phosphorylase/metabolism
13.
Biochemistry ; 51(34): 6715-7, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22870934

ABSTRACT

The transition state for the Trypanosoma cruzi uridine phosphorylase (TcUP) reaction has an expanded S(N)2 character. We used binding isotope effects (BIE's) to probe uridine distortion in the complex with TcUP and sulfate to mimic the Michaelis complex. Inverse 1'-(3)H and 5'-(3)H BIE's indicate a constrained bonding environment of these groups in the complex. Quantum chemical modeling identified a uridine conformer whose calculated BIE's match the experimental values. This conformer differs in sugar pucker and uracil orientation from the unbound conformer and the transition-state structure. These results support ground-state stabilization in the Michaelis complex.


Subject(s)
Protozoan Proteins/chemistry , Trypanosoma cruzi/enzymology , Uridine Phosphorylase/chemistry , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/metabolism , Isotope Labeling , Kinetics , Models, Chemical , Protein Binding , Protozoan Proteins/metabolism , Tritium/chemistry , Tritium/metabolism , Trypanosoma cruzi/chemistry , Uridine/chemistry , Uridine/metabolism , Uridine Phosphorylase/metabolism
14.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 8): 968-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22868762

ABSTRACT

Uridine phosphorylase (UPh), which is a key enzyme in the reutilization pathway of pyrimidine nucleoside metabolism, is a validated target for the treatment of infectious diseases and cancer. A detailed analysis of the interactions of UPh with the therapeutic ligand 5-fluorouracil (5-FUra) is important for the rational design of pharmacological inhibitors of these enzymes in prokaryotes and eukaryotes. Expanding on the preliminary analysis of the spatial organization of the active centre of UPh from the pathogenic bacterium Salmonella typhimurium (StUPh) in complex with 5-FUra [Lashkov et al. (2009), Acta Cryst. F65, 601-603], the X-ray structure of the StUPh-5-FUra complex was analysed at atomic resolution and an in silico model of the complex formed by the drug with UPh from Vibrio cholerae (VchUPh) was generated. These results should be considered in the design of selective inhibitors of UPhs from various species.


Subject(s)
Fluorouracil/pharmacology , Salmonella typhimurium/enzymology , Uridine Phosphorylase/chemistry , Vibrio cholerae/enzymology , Catalysis , Catalytic Domain , Cluster Analysis , Enzyme Inhibitors/pharmacology , Ligands , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Solvents
15.
FEBS Lett ; 586(7): 1044-8, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22569261

ABSTRACT

Escherichia coli YrhB (10.6 kDa) from strain BL21(DE3) that is commonly used for protein overexpression is a stable chaperone-like protein and indispensable for supporting the growth of BL21(DE3) at 48 °C but not defined as conventional heat shock protein (HSP). YrhB effectively prevented heat-induced aggregation of ribonucleotide synthetase (PurK). Without ATP, YrhB alone promoted in vitro refolding of uridine phosphorylase (UDP) and protected thermal denaturation of the refolded UDP. As a cis-acting fusion partner, YrhB also significantly reduced inclusion body formation of nine aggregation-prone heterologous proteins in BL21(DE3). Unlike conventional small HSPs, YrhB remained monomer under heat shock condition.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Molecular Chaperones/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Expression Regulation, Enzymologic , Heat-Shock Response , Hot Temperature , Inclusion Bodies , Kinetics , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Weight , Mutation , Promoter Regions, Genetic , Protein Denaturation , Protein Refolding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sigma Factor/genetics , Sigma Factor/metabolism , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/metabolism
16.
Biochemistry ; 50(42): 9158-66, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21932786

ABSTRACT

The reversible phosphorolysis of uridine to generate uracil and ribose 1-phosphate is catalyzed by uridine phosphorylase and is involved in the pyrimidine salvage pathway. We define the reaction mechanism of uridine phosphorylase from Trypanosoma cruzi by steady-state and pre-steady-state kinetics, pH-rate profiles, kinetic isotope effects from uridine, and solvent deuterium isotope effects. Initial rate and product inhibition patterns suggest a steady-state random kinetic mechanism. Pre-steady-state kinetics indicated no rate-limiting step after formation of the enzyme-products ternary complex, as no burst in product formation is observed. The limiting single-turnover rate constant equals the steady-state turnover number; thus, chemistry is partially or fully rate limiting. Kinetic isotope effects with [1'-(3)H]-, [1'-(14)C]-, and [5'-(14)C,1,3-(15)N(2)]uridine gave experimental values of (α-T)(V/K)(uridine) = 1.063, (14)(V/K)(uridine) = 1.069, and (15,ß-15)(V/K)(uridine) = 1.018, in agreement with an A(N)D(N) (S(N)2) mechanism where chemistry contributes significantly to the overall rate-limiting step of the reaction. Density functional theory modeling of the reaction in gas phase supports an A(N)D(N) mechanism. Solvent deuterium kinetic isotope effects were unity, indicating that no kinetically significant proton transfer step is involved at the transition state. In this N-ribosyl transferase, proton transfer to neutralize the leaving group is not part of transition state formation, consistent with an enzyme-stabilized anionic uracil as the leaving group. Kinetic analysis as a function of pH indicates one protonated group essential for catalysis and for substrate binding.


Subject(s)
Trypanosoma cruzi/enzymology , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/metabolism , Animals , Catalysis , Deuterium Exchange Measurement , Hydrogen-Ion Concentration , Phosphorylation , Ribosemonophosphates/chemistry , Substrate Specificity , Uracil/chemistry , Uridine/chemistry
17.
J Struct Biol ; 176(2): 229-37, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21855639

ABSTRACT

Uridine phosphorylase (UPP) catalyzes the reversible conversion of uridine to uracil and ribose-1-phosphate and plays an important pharmacological role in activating fluoropyrimidine nucleoside chemotherapeutic agents such as 5-fluorouracil and capecitabine. Most vertebrate animals, including humans, possess two homologs of this enzyme (UPP1 & UPP2), of which UPP1 has been more thoroughly studied and is better characterized. Here, we report two crystallographic structures of human UPP2 (hUPP2) in distinctly active and inactive conformations. These structures reveal that a conditional intramolecular disulfide bridge can form within the protein that dislocates a critical phosphate-coordinating arginine residue (R100) away from the active site, disabling the enzyme. In vitro activity measurements on both recombinant hUPP2 and native mouse UPP2 confirm the redox sensitivity of this enzyme, in contrast to UPP1. Sequence analysis shows that this feature is conserved among UPP2 homologs and lacking in all UPP1 proteins due to the absence of a necessary cysteine residue. The state of the disulfide bridge has further structural consequences for one face of the enzyme that suggest UPP2 may have additional functions in sensing and initiating cellular responses to oxidative stress. The molecular details surrounding these dynamic aspects of hUPP2 structure and regulation provide new insights as to how novel inhibitors of this protein may be developed with improved specificity and affinity. As uridine is emerging as a promising protective compound in neuro-degenerative diseases, including Alzheimer's and Parkinson's, understanding the regulatory mechanisms underlying UPP control of uridine concentration is key to improving clinical outcomes in these illnesses.


Subject(s)
Uridine Phosphorylase/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Cystine/chemistry , Enzyme Assays , Humans , Hydrogen Bonding , Mice , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Uracil/analogs & derivatives , Uracil/chemistry , Uridine Phosphorylase/antagonists & inhibitors
18.
Biochemistry ; 50(30): 6549-58, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21707079

ABSTRACT

Uridine phosphorylase (UP), a key enzyme in the pyrimidine salvage pathway, catalyzes the reversible phosphorolysis of uridine or 2'-deoxyuridine to uracil and ribose 1-phosphate or 2'-deoxyribose 1-phosphate. This enzyme belongs to the nucleoside phosphorylase I superfamily whose members show diverse specificity for nucleoside substrates. Phylogenetic analysis shows Streptococcus pyogenes uridine phosphorylase (SpUP) is found in a distinct branch of the pyrimidine subfamily of nucleoside phosphorylases. To further characterize SpUP, we determined the crystal structure in complex with the products, ribose 1-phosphate and uracil, at 1.8 Å resolution. Like Escherichia coli UP (EcUP), the biological unit of SpUP is a hexamer with an α/ß monomeric fold. A novel feature of the active site is the presence of His169, which structurally aligns with Arg168 of the EcUP structure. A second active site residue, Lys162, is not present in previously determined UP structures and interacts with O2 of uracil. Biochemical studies of wild-type SpUP showed that its substrate specificity is similar to that of EcUP, while EcUP is ∼7-fold more efficient than SpUP. Biochemical studies of SpUP mutants showed that mutations of His169 reduced activity, while mutation of Lys162 abolished all activity, suggesting that the negative charge in the transition state resides mostly on uracil O2. This is in contrast to EcUP for which transition state stabilization occurs mostly at O4.


Subject(s)
Bacterial Proteins/chemistry , Multigene Family , Streptococcus pyogenes/enzymology , Uridine Phosphorylase/chemistry , Bacterial Proteins/genetics , Catalysis , Catalytic Domain/genetics , Crystallography, X-Ray , Mutagenesis, Site-Directed , Protein Folding , Ribosemonophosphates/chemistry , Static Electricity , Substrate Specificity/genetics , Uracil/chemistry , Uridine Phosphorylase/genetics
19.
J Am Chem Soc ; 133(25): 9923-31, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21599004

ABSTRACT

Uridine phosphorylase catalyzes the reversible phosphorolysis of uridine and 2'-deoxyuridine to generate uracil and (2-deoxy)ribose 1-phosphate, an important step in the pyrimidine salvage pathway. The coding sequence annotated as a putative nucleoside phosphorylase in the Trypanosoma cruzi genome was overexpressed in Escherichia coli , purified to homogeneity, and shown to be a homodimeric uridine phosphorylase, with similar specificity for uridine and 2'-deoxyuridine and undetectable activity toward thymidine and purine nucleosides. Competitive kinetic isotope effects (KIEs) were measured and corrected for a forward commitment factor using arsenate as the nucleophile. The intrinsic KIEs are: 1'-(14)C = 1.103, 1,3-(15)N(2) = 1.034, 3-(15)N = 1.004, 1-(15)N = 1.030, 1'-(3)H = 1.132, 2'-(2)H = 1.086, and 5'-(3)H(2) = 1.041 for this reaction. Density functional theory was employed to quantitatively interpret the KIEs in terms of transition-state structure and geometry. Matching of experimental KIEs to proposed transition-state structures suggests an almost synchronous, S(N)2-like transition-state model, in which the ribosyl moiety possesses significant bond order to both nucleophile and leaving groups. Natural bond orbital analysis allowed a comparison of the charge distribution pattern between the ground-state and the transition-state models.


Subject(s)
Arsenates/pharmacology , Biocatalysis , Trypanosoma cruzi/enzymology , Uridine Phosphorylase/metabolism , Uridine/metabolism , Cloning, Molecular/methods , Kinetics , Models, Molecular , Static Electricity , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/genetics
20.
Anticancer Res ; 31(3): 831-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21498703

ABSTRACT

BACKGROUND: In order to reduce toxicity and to enhance anticancer activity of nitrogen mustards, three hybrid steroidal esters were synthesized and tested in vitro against human pancreatic cancer cells expressing uridine phosphorylase (UPase). The inhibition potency against a target protein implicated in the chemotherapy of solid tumors, such as UPase, is of fundamental importance in the design and synthesis of new anticancer drugs. MATERIALS AND METHODS: MTT colorimetric assay and molecular docking were employed for the in vitro and in silico drug evaluation, respectively. RESULTS: A difference in cell sensitivity was found, which followed the known different UPase expression in the cell lines. Molecular docking studies on UPase protein, revealed the tested compounds to be bound to the binding cavity of the protein, with different affinity. Between the two D-modified compounds, the D-homo-aza (lactam)-hybrid compound (C2) was found to interact with the protein in a more efficient way. CONCLUSION: The molecular docking data were in accordance with the in vitro results, where the lactam steroid alkylator showed significantly higher cytostatic and cytotoxic activity than the non-D-modified compounds, which also correlated with the level of UPase expression in the pancreatic cancer cells.


Subject(s)
Alkylating Agents/pharmacology , Antineoplastic Agents/pharmacology , Computational Biology , Molecular Targeted Therapy , Steroids/pharmacology , Uridine Phosphorylase/antagonists & inhibitors , Alkylating Agents/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Fluorouracil/chemistry , Fluorouracil/metabolism , Humans , Ligands , Models, Molecular , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Steroids/chemistry , Thermodynamics , Thiouracil/analogs & derivatives , Thiouracil/chemistry , Thiouracil/metabolism , Uridine Phosphorylase/chemistry , Uridine Phosphorylase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...