Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Agric Food Chem ; 68(34): 9188-9194, 2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32806118

RESUMEN

A rapid in vitro enzymatic biosynthesis system has been developed as a biological manufacturing platform with potential industrial uses. Cytidine 5'-monophosphate (5'-CMP) is a key intermediate in the preparation of several nucleotide derivatives and is widely used in food and pharmaceutical industries. In this study, a highly efficient biosynthesis system was constructed for manufacturing 5'-CMP in vitro. Cytidine kinase (CK) was used for the biotransformation of cytidine to 5'-CMP, while polyphosphate kinase (PPK) was coupled for adenosine triphosphate regeneration. Both CK and PPK were selected from extremophiles, possessing great potential for biocatalytic synthesis. The effects of temperature, substrate concentration, and enzyme ratios were investigated to enhance the titer and yield of 5'-CMP. After optimization, 96 mM 5'-CMP was produced within 6 h, and the yield reached nearly 100%. This work highlights the ease of 5'-CMP production by an in vitro biomanufacturing platform and provides a green and efficient approach for the industrial synthesis of 5'-CMP.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas/metabolismo , Citidina Monofosfato/biosíntesis , Extremófilos/metabolismo , Secuencia de Aminoácidos , Bacterias/química , Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biotransformación , Citidina Monofosfato/química , Estabilidad de Enzimas , Extremófilos/química , Extremófilos/enzimología , Extremófilos/genética , Fosfotransferasas (Aceptor del Grupo Fosfato)/química , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Alineación de Secuencia , Uridina Quinasa/química , Uridina Quinasa/genética , Uridina Quinasa/metabolismo
2.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 613-618, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27906629

RESUMEN

Uridine-cytidine kinase (UCK) catalyzes the phosphorylation of uridine, cytidine, and several pyrimidine ribonucleoside analogs. We overexpressed and purified the two known isoforms of human UCK in Escherichia coli, produced a specific antibody against UCK1 and characterized the kinetic properties of UCK1 and 2. The Vmax of purified recombinant UCK2 was 22- and 8-fold higher with uridine and cytidine, respectively, compared to those observed for the purified recombinant UCK1 enzyme. The Km of UCK1 was 39- and 40-fold higher with uridine and cytidine, respectively, compared to those observed for the purified recombinant UCK2 enzyme. The UCK1 antibody showed no cross reactivity against UCK2. Our data showed that UCK1 and 2 are both expressed in several neuroblastoma cell lines, including four MYCN single copy cell lines and five MYCN amplified cell lines, with the exception that UCK1 was not expressed in SJNB8. These results indicate that UCK2 in neuroblastoma might be used as a selective target for chemotherapy using UCK2-dependent pyrimidine analogues.


Asunto(s)
Nucleósido-Fosfato Quinasa/genética , Uridina Quinasa/genética , Adenosina Trifosfato/química , Citidina/química , Escherichia coli , Expresión Génica , Humanos , Cinética , Neuroblastoma/enzimología , Nucleósido-Fosfato Quinasa/biosíntesis , Nucleósido-Fosfato Quinasa/química , Especificidad por Sustrato , Uridina/química , Uridina Quinasa/biosíntesis , Uridina Quinasa/química
3.
Molecules ; 21(4): 417, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-27070566

RESUMEN

Uridine-cytidine kinase 2 is implicated in uncontrolled proliferation of abnormal cells and it is a hallmark of cancer, therefore, there is need for effective inhibitors of this key enzyme. In this study, we employed the used of in silico studies to find effective UCK2 inhibitors of natural origin using bioinformatics tools. An in vitro kinase assay was established by measuring the amount of ADP production in the presence of ATP and 5-fluorouridine as a substrate. Molecular docking studies revealed an interesting ligand interaction with the UCK2 protein for both flavokawain B and alpinetin. Both compounds were found to reduce ADP production, possibly by inhibiting UCK2 activity in vitro. In conclusion, we have identified flavokawain B and alpinetin as potential natural UCK2 inhibitors as determined by their interactions with UCK2 protein using in silico molecular docking studies. This can provide information to identify lead candidates for further drug design and development.


Asunto(s)
Inhibidores Enzimáticos/química , Flavanonas/química , Flavonoides/química , Uridina Quinasa/química , Adenosina Difosfato/biosíntesis , Alpinia/enzimología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Simulación por Computador , Inhibidores Enzimáticos/uso terapéutico , Flavanonas/uso terapéutico , Flavonoides/uso terapéutico , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Rizoma/enzimología , Uridina Quinasa/antagonistas & inhibidores
4.
Protein J ; 34(6): 411-20, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26510656

RESUMEN

Uridine-cytidine kinase catalyzes phosphorylation of the pyrimidine nucleosides uridine and cytidine and plays an important role in nucleotide metabolism. However, the detailed molecular mechanism of these reactions remains to be elucidated. Here, we determined the structure of the ternary complex of Uridine-cytidine kinase from Thermus thermophilus HB8 with both cytidine and ß,γ-methyleneadenosine 5'-triphosphate, a non-hydrolysable ATP analogue. Substrate binding is accompanied by substantial domain movement that allows the substrate-binding cleft to close. The terminal phosphodiester bond of the ATP analogue is in an ideal location for an inline attack of the 5'-hydroxyl group of cytidine. Asp40 is located near the 5'-hydroxyl group of cytidine. Mutation of this conserved residue to Asn or Ala resulted in a complete loss of enzyme activity, which is consistent with the notion that Asp40 acts as a general base that activates the 5'-hydroxyl group of cytidine. The pH profile of the activity showed an apparent pK a value of 7.4. Based on this structure, a likely mechanism of the catalytic step is discussed.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Thermus thermophilus/enzimología , Uridina Quinasa/química , Uridina Quinasa/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Concentración de Iones de Hidrógeno , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Nucleósidos de Pirimidina , Alineación de Secuencia , Thermus thermophilus/genética , Uridina Quinasa/genética
5.
Org Biomol Chem ; 7(13): 2716-24, 2009 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-19532987

RESUMEN

The mechanisms of enzyme-catalyzed phosphate transfer and hydrolysis reactions are of great interest due to their importance and abundance in biochemistry. The reaction may proceed in a stepwise fashion, with either a pentavalent phosphorane or a metaphosphate anion intermediate, or by a concerted SN2 mechanism. Despite much theoretical work focused on a few key enzymes, a consensus for the mechanism has not been reached, and examples of all three possibilities have been demonstrated. We have investigated the mechanism of human uridine-cytidine kinase 2 (UCK2, EC 2.7.1.48), which catalyzes the transfer of a phosphate group from ATP to the ribose 5'-hydroxyl of cytidine and uridine. UCK2 is normally expressed in human placenta, but is overexpressed in certain cancer cells, where it is responsible for activating a class of antitumor prodrugs. The UCK2 mechanism was investigated by generating a 2D potential energy surface as a function of the P-O bonds forming and breaking, with energies calculated using a quantum mechanics/molecular mechanics potential (B3LYP/6-31G(d):AMBER). The mechanism of phosphate transfer is shown to be concerted, and is accompanied by concerted proton transfer from the 5'-hydroxyl to a conserved active site aspartic acid that serves as a catalytic base. The calculated barrier for this reaction is 15.1 kcal/mol, in relatively good agreement with the experimental barrier of 17.5 kcal/mol. The interactions of the enzyme active site with the reactant, transition state, and product are examined for their implications on the design of anticancer prodrugs or positron emission tomography (PET) reporter probes for this enzyme.


Asunto(s)
Fosfatos/química , Fosfatos/metabolismo , Teoría Cuántica , Termodinámica , Uridina Quinasa/química , Uridina Quinasa/metabolismo , Catálisis , Humanos , Hidrólisis , Modelos Moleculares , Estructura Molecular
6.
Parasitology ; 136(6): 595-602, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19368743

RESUMEN

Entamoeba histolytica is an intestinal parasite and the causative agent of amoebiasis, which is a significant source of morbidity and mortality in developing countries. Although anti-amoebic drugs such as metronidazole, emetine, chloroquine and nitazoxanide are generally effective, there is always potential for development of drug resistance. In order to find novel targets to control E. histolytica proliferation we cloned, expressed and purified thymidine kinase (Eh-TK) and uridine-cytidine kinase (Eh-UCK) from E. histolytica. Eh-TK phosphorylates thymidine with a Km of 0.27 microm, whereas Eh-UCK phosphorylates uridine and cytidine with Km of 0.74 and 0.22 mM, respectively. For both enzymes, ATP acts as specific phosphate donor. In order to find alternative treatments of E. histolytica infection we tested numerous nucleoside analogues and related compounds as inhibitors and/or substrates of Eh-TK and Eh-UCK, and active compounds against E. histolytica in cell culture. Our results indicate that inhibitors or alternative substrates of the enzymes, although partially reducing protozoan proliferation, are reversible and not likely to become drugs against E. histolytica infections.


Asunto(s)
Antiprotozoarios/farmacología , Entamoeba histolytica/efectos de los fármacos , Entamoeba histolytica/enzimología , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Uridina Quinasa/genética , Uridina Quinasa/metabolismo , Secuencia de Aminoácidos , Animales , Proliferación Celular , Clonación Molecular , Entamoeba histolytica/citología , Entamoeba histolytica/genética , Humanos , Datos de Secuencia Molecular , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Timidina Quinasa/antagonistas & inhibidores , Timidina Quinasa/química , Uridina Quinasa/antagonistas & inhibidores , Uridina Quinasa/química
7.
J Biol Chem ; 283(37): 25238-25246, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18591240

RESUMEN

Pseudouridine, a non-classical nucleoside present in human urine as a degradation product of RNAs, is one of the few molecules that has a glycosidic C-C bond. Through a data base mining approach involving transcriptomic data, we have molecularly identified two enzymes that are involved in the metabolism of pseudouridine in uropathogenic Escherichia coli, the principal agent of urinary tract infections in humans. The first enzyme, coded by the gene yeiC, specifically phosphorylates pseudouridine to pseudouridine 5'-phosphate. Accordingly, yeiC(-) mutants are unable to metabolize pseudouridine, in contrast to wild-type E. coli UTI89. The second enzyme, encoded by the gene yeiN belonging to the same operon as yeiC, catalyzes the conversion of pseudouridine 5'-phosphate to uracil and ribose 5-phosphate in a divalent cation-dependent manner. Remarkably, the glycosidic C-C bond of pseudouridine is cleaved in the course of this reaction, indicating that YeiN is the first molecularly identified enzyme able to hydrolyze a glycosidic C-C bond. Though this reaction is easily reversible, the association of YeiN with pseudouridine kinase indicates that it serves physiologically to metabolize pseudouridine 5'-phosphate rather than to form it. YeiN is homologous to Thermotoga maritima IndA, a protein with a new fold, which we now show to act also as a pseudouridine-5'-phosphate glycosidase. Data base mining indicates that most eukaryotes possess homologues of pseudouridine kinase and pseudouridine-5'-phosphate glycosidase and that these are most often associated in a single bifunctional protein. The gene encoding this bifunctional protein is absent from the genomes of man and other mammals, indicating that the capacity for metabolizing pseudouridine has been lost late in evolution.


Asunto(s)
Escherichia coli/metabolismo , Seudouridina/química , ARN/química , Secuencia de Aminoácidos , Glicósido Hidrolasas/química , Glicósidos/química , Humanos , Modelos Químicos , Modelos Genéticos , Datos de Secuencia Molecular , Nucleósidos/química , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Homología de Secuencia de Aminoácido , Uridina Quinasa/química
8.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 3): 278-84, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15735337

RESUMEN

Uridine-cytidine nucleoside kinase 2 (UCK2) is the rate-limiting enzyme in the pyrimidine-nucleotide salvage pathway. UCK2 catalyzes the phosphorylation of the natural ribonucleosides cytidine and uridine to cytidine 5'-monophosphate (CMP) and uridine 5'-monophosphate (UMP), respectively, and activates several important frontline antimetabolite drugs. The present contribution reports the rapid crystal structure determination of human UCK2 complexed with a magnesium ion and the reaction products adenosine 5'-diphosphate (ADP) and CMP. Diffraction data were collected on a copper rotating-anode X-ray generator from one native UCK2 crystal and a single samarium-derivative crystal. Utilizing the relatively high anomalous signal from the samarium derivative at the Cu Kalpha wavelength, the structure was determined by single isomorphous replacement and single anomalous signal (SIRAS) phasing techniques. Two of the four major samarium sites are located in the active sites of the two UCK2 molecules that form the asymmetric unit and appear to displace the magnesium ions present in the native crystals. The crystal structures of UCK2 alone and in complex with various ligands have recently been determined using traditional multiple isomorphous replacement (MIR) phasing techniques and data from three heavy-atom derivatives. The reported structures validate our independently determined structure. Of more than 1000 kinase crystal structure entries in the Protein Data Bank, less than 1% of them have been determined by SIRAS. For the published kinase crystal structures determined by SIRAS, all data were reportedly collected at various synchrotron-radiation facilities. This study demonstrates that diffraction data collected from a single samarium derivative using Cu Kalpha radiation provides sufficient phasing power to determine a novel macromolecular crystal structure.


Asunto(s)
Samario/química , Uridina Quinasa/química , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica
9.
Structure ; 12(5): 751-64, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15130468

RESUMEN

Uridine-cytidine kinase (UCK) catalyzes the phosphorylation of uridine and cytidine and activates pharmacological ribonucleoside analogs. Here we present the crystal structures of human UCK alone and in complexes with a substrate, cytidine, a feedback inhibitor, CTP or UTP, and with phosphorylation products, CMP and ADP, respectively. Free UCK takes an alpha/beta mononucleotide binding fold and exists as a homotetramer with 222 symmetry. Upon inhibitor binding, one loop region was loosened, causing the UCK tetramer to be distorted. Upon cytidine binding, a large induced fit was observed at the uridine/cytidine binding site, which endows UCK with a strict specificity for pyrimidine ribonucleosides. The first UCK structure provided the structural basis for the specificity, catalysis, and regulation of human uridine-cytidine kinase, which give clues for the design of novel antitumor and antiviral ribonucleoside analogs that inhibit RNA synthesis.


Asunto(s)
Uridina Quinasa/química , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Retroalimentación Fisiológica/fisiología , Humanos , Ligandos , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Especificidad por Sustrato , Ultracentrifugación , Uridina Quinasa/antagonistas & inhibidores , Uridina Quinasa/metabolismo
10.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 8): 1477-8, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12876357

RESUMEN

Uridine-cytidine kinase (UCK), which converts uridine and cytidine to their corresponding monophosphates, is a rate-limiting enzyme involved in the salvage pathway of pyrimidine-nucleotide biosynthesis. Two members of human UCK, named UCK1 and UCK2, were cloned recently and UCK2 was reported to play a crucial role in activating anti-tumour pro-drugs in human cancer cells. Human UCK2 was expressed, purified and crystallized alone or in complex with various ligands. Free UCK and UCK complexes were crystallized in six crystal forms. Form I (ligand-free) belongs to space group P2(1)2(1)2, with unit-cell parameters a = 83.1, b = 93.7, c = 157.1 A. Forms IIa (with CTP), IIb (with UTP) and IIc (with cytidine) belong to space group F222, with unit-cell parameters a = 133.3, b = 247.3, c = 91.6 A (IIa), a = 132.1, b = 247.0, c = 91.5 A (IIb) and a = 136.7, b = 246.3, c = 90.4 A (IIc), respectively. Form III (with ATPgammaS) belongs to space group C222(1), with unit-cell parameters a = 70.3, b = 149.9, c = 117.2 A. Form IV (with cytidine and ATP) belongs to space group C2, with unit-cell parameters a = 89.0, b = 109.7, c = 64.8 A, beta = 95.3 degrees. Diffraction data were collected from these crystals; form IV diffracted to 1.8 A resolution.


Asunto(s)
Uridina Quinasa/química , Catálisis , Cristalización , Cristalografía por Rayos X/métodos , Escherichia coli/metabolismo , Humanos , Ligandos , Estructura Terciaria de Proteína
11.
BMC Cell Biol ; 3: 23, 2002 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-12199906

RESUMEN

BACKGROUND: Epstein-Barr virus (EBV) infects resting B-lymphocytes and transforms them into immortal proliferating lymphoblastoid cell lines (LCLs) in vitro. The transformed immunoblasts may grow up as immunoblastic lymphomas in immuno-suppressed hosts. RESULTS: In order to identify cellular protein targets that may be involved in Epstein-Barr virus mediated B-cell transformation, human LCL cDNA library was screened with one of the transformation associated nuclear antigens, EBNA-3 (also called EBNA-3A), using the yeast two-hybrid system. A clone encoding a fragment of a novel human protein was isolated (clone 538). The interaction was confirmed using in vitro binding assays. A full-length cDNA clone (F538) was isolated. Sequence alignment with known proteins and 3D structure predictions suggest that F538 is a novel human uridine kinase/uracil phosphoribosyltransferase. The GFP-F538 fluorescent fusion protein showed a preferentially cytoplasmic distribution but translocated to the nucleus upon co-expression of EBNA-3. A naturally occurring splice variant of F538, that lacks the C-terminal uracil phosphoribosyltransferase part but maintain uridine kinase domain, did not translocate to the nucleus in the presence of EBNA3. Antibody that was raised against the bacterially produced GST-538 protein showed cytoplasmic staining in EBV negative Burkitt lymphomas but gave a predominantly nuclear staining in EBV positive LCL-s and stable transfected cells expressing EBNA-3. CONCLUSION: We suggest that EBNA-3 by direct protein-protein interaction induces the nuclear accumulation of a novel enzyme, that is part of the ribonucleotide salvage pathway. Increased intranuclear levels of UK/UPRT may contribute to the metabolic build-up that is needed for blast transformation and rapid proliferation.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Proteínas Nucleares/metabolismo , Pentosiltransferasa/metabolismo , Uridina Quinasa/metabolismo , Secuencia de Aminoácidos/genética , Animales , Proteínas de Caenorhabditis elegans , Cryptosporidium parvum , Dimerización , Herpesvirus Humano 4/genética , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Pentosiltransferasa/química , Pentosiltransferasa/genética , Unión Proteica/genética , Mapeo de Interacción de Proteínas/métodos , Estructura Cuaternaria de Proteína/genética , Estructura Secundaria de Proteína/genética , Proteínas Protozoarias , Alineación de Secuencia/métodos , Uridina Quinasa/química , Uridina Quinasa/genética
12.
Int Microbiol ; 2(1): 3-10, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10943384

RESUMEN

Carbon dioxide fixation is a polyphyletic trait that has evolved in widely separated prokaryotic branches. The three principal CO2-assimilation pathways are (i) the reductive pentose-phosphate cycle, i.e. the Calvin-Benson cycle; (ii) the reductive citric acid (or Arnon) cycle; and (iii) the net synthesis of acetyl-CoA from CO/CO2, or Wood pathway. Sequence analysis and the comparative biochemistry of these routes suggest that all of them were shaped to a considerable extent by the evolutionary recruitment of enzymes. Molecular phylogenetic trees show that the Calvin-Benson cycle was a relatively late development in the (eu)bacterial branch, suggesting that some form(s) of carbon assimilation may have been operative before chlorophyll-based photosynthesis. On the other hand, the ample phylogenetic distribution of both the Arnon and the Wood pathways does not allow us to infer which one of them is older. However, different lines of evidence, including experimental reports on the NiS/FeS-mediated C-C bond formation from CO and CH3SH are used here to argue that the first CO2-fixation route may have been a semi-enzymatic Wood-like pathway.


Asunto(s)
Evolución Biológica , Dióxido de Carbono/metabolismo , Metabolismo Energético , Origen de la Vida , Acetilcoenzima A/biosíntesis , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Ciclo del Ácido Cítrico , Evolución Molecular , Datos de Secuencia Molecular , Vía de Pentosa Fosfato , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Filogenia , Células Procariotas/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Uridina Quinasa/química
13.
FEMS Microbiol Lett ; 151(2): 257-61, 1997 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9228761

RESUMEN

The 621 bp udk gene encoding Borrelia burgdorferi potential uridine kinase, involved in the pyrimidine salvage pathway, was cloned and sequenced. The B burgdorferi protein has a molecular mass of 24 kDa in sodium dodecyl sulfate-polyacrylamide gel. The N-terminal sequence of the protein, Ala-Lys-Ile-Ile, is identical to that predicted but lacks N-terminal methionine. udk is located at around 15 kb from the left telomere and forms an operon with an upstream ORF. A likely hypothesis for the role of the pyrimidine salvage pathway is the sole use of endogenous nucleotides for Borrelia.


Asunto(s)
Grupo Borrelia Burgdorferi/enzimología , Pirimidinas/metabolismo , Uridina Quinasa/genética , Secuencia de Aminoácidos , Grupo Borrelia Burgdorferi/genética , Clonación Molecular , Datos de Secuencia Molecular , Operón , Uridina Quinasa/química , Uridina Quinasa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...