Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Arch Biochem Biophys ; 362(2): 339-45, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9989944

ABSTRACT

The enzyme that catalyzes the formation of GDP-d-mannose from GTP and alpha-d-mannose-1-P was purified about 2300-fold to near homogeneity from the soluble fraction of Mycobacterium smegmatis. At the final stage of purification, a major protein band of 37 kDa was observed and this band was specifically labeled, and in a concentration-dependent manner, by the photoaffinity probe 8-N3-GDP[32P]-d-mannose. The purified enzyme was stable for several months when kept in the frozen state. The 37-kDa band was subjected to protein sequencing and one peptide sequence of 25 amino acids showed over 80% identity to GDP-mannose pyrophosphorylases of pig liver and Saccharomyces cerevesiae. In contrast to some other bacterial GDP-mannose pyrophosphorylases, the mycobacterial enzyme was not multifunctional and did not have phosphomannose isomerase or phosphoglucose isomerase activity. Also, in contrast to the pig liver enzyme which uses mannose-1-P or glucose-1-P plus GTP to synthesize either GDP-mannose or GDP-glucose, the mycobacterial enzyme was specific for mannose-1-P as the sugar phosphate substrate. The enzyme was also relatively specific for GTP as the nucleoside triphosphate substrate. ITP was about 18% as effective as GTP, but ATP, CTP, and UTP were inactive. The activity of the enzyme was inhibited by GDP-glucose and glucose-1-P, although neither was a substrate for this enzyme. The pH optimum for the enzyme was 8.0, and Mg2+ was the best cation with optimum activity at about 5 mM. This enzyme is important for producing the activated form of mannose for formation of cell wall lipoarabinomannan and various mannose-containing glycolipids and polysaccharides.


Subject(s)
Mycobacterium smegmatis/enzymology , Nucleotidyltransferases/metabolism , Amino Acid Sequence , Animals , Enzyme Stability , Glucosephosphates/pharmacology , Guanosine Diphosphate Mannose/analogs & derivatives , Guanosine Diphosphate Mannose/metabolism , Guanosine Diphosphate Sugars/pharmacology , Guanosine Triphosphate/metabolism , Hydrogen-Ion Concentration , Inosine Triphosphate/metabolism , Kinetics , Magnesium/pharmacology , Mannose-6-Phosphate Isomerase/metabolism , Mannosephosphates/metabolism , Molecular Sequence Data , Molecular Weight , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/isolation & purification , Photoaffinity Labels , Sequence Analysis , Sequence Homology, Amino Acid , Substrate Specificity
2.
Mol Pharmacol ; 49(2): 360-4, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8632770

ABSTRACT

Microinjection, but not extracellular application, of cytidine-5'-diphosphate-D-glucose (CDPG) has been shown to elicit Ca(2+)-dependent currents in Xenopus laevis oocytes. These responses were comparable to those of inositol-1,4,5-trisphosphate (InsP3) in being both rapid and dose dependent. For example, maximal amplitudes of CDPG-induced current were similar (approximately 365 +/- 75 nA at 1 microM CDPG) to those of InsP3. The CDPG currents were insensitive to removal of extracellular Ca2+, indicating the dependence on Ca2+ release from intracellular Ca2+ stores but not on Ca2+ entry through plasma membrane. CDPG-induced currents were reduced or abolished by pretreatment with thapsigargin, by injection of the Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, or by extracellular perfusion of the Cl- channel blocker niflumic acid but were insensitive to injection of the InsP3 antagonist heparin. These results suggest that CDPG induces Ca2+ discharge from intracellular Ca2+ stores via a mechanism distinct from that of InsP3 in Xenopus oocytes. Another pyrimidine nucleotide-glucose derivative, uridine-5'-diphosphate-alpha-D-glucose, also induced Ca(2+)-dependent currents, but the activity was lower than that of CDPG (maximal amplitude, 272 +/- 62 nA). Other nucleotide-glucose compounds (adenosine-5'-diphosphate-D-glucose, guanosine-5'-diphosphate-D-glucose, and thymidine-5'-diphosphate-D-glucose) had no current responses when injected into oocytes. After injection of CDPG, CDPG-induced Ca2+ release appeared to couple to a Ca2+ entry pathway similar to that coupled to InsP3. These results indicate that pyrimidine nucleotide-glucose conjugates may provide novel pharmacological tools for the study of Ca2+ signaling in oocytes.


Subject(s)
Calcium/pharmacology , Cell Membrane/physiology , Inositol 1,4,5-Trisphosphate/pharmacology , Nucleoside Diphosphate Sugars/pharmacology , Oocytes/physiology , Adenosine Diphosphate Glucose/pharmacology , Animals , Cell Membrane/drug effects , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , Glucose/analogs & derivatives , Glucose/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Guanosine Diphosphate Sugars/pharmacology , Inositol 1,4,5-Trisphosphate/administration & dosage , Kinetics , Membrane Potentials/drug effects , Microinjections , Oocytes/drug effects , Sulfides , Thiophenes , Uridine Diphosphate Glucose/pharmacology , Xenopus
3.
FEBS Lett ; 243(2): 413-6, 1989 Jan 30.
Article in English | MEDLINE | ID: mdl-2917659

ABSTRACT

Guanosine diphosphate (GDP) esters of 2-deoxy-D-glucose (2dGlc), 2-deoxy-2-fluoro-D-mannose (2FMan), 3-deoxy-D-mannose (3dMan), 4-deoxy-D-mannose (4dMan) and 6-deoxy-D-mannose (6dMan) have been synthesised and tested for their ability to act as inhibitors of dolichyl phosphate mannose synthesis (enzyme: GDP-mannose:dolichyl-phosphate mannosyltransferase, EC 2.4.1.83) in chick embryo cell microsomal membranes. The following order of efficiency was found with the apparent Ki in parentheses: GDP-6dMan (0.40 microM +/- 0.15) greater than GDP-3dMan (1.0 microM +/- 0.1) = GDP-2dGlc (1.3 microM +/- 0.2) greater than GDP-4dMan (3.1 microM +/- 0.1) GDP-2FMan (15 microM +/- 0). For comparison the Km for GDP-Man was 0.52 microM +/- 0.02 and the Ki for GDP was 56 microM +/- 2. These results indicate that the 6-hydroxyl group of mannose is not crucial for enzyme-substrate recognition, whereas the 2- and 3-hydroxyls may have some involvement. The 4-hydroxyl appears to be an important determinant for enzyme-substrate recognition in this mannosyltransferase.


Subject(s)
Guanosine Diphosphate Sugars/metabolism , Hexosephosphates/metabolism , Hexosyltransferases/metabolism , Mannosephosphates/metabolism , Mannosyltransferases/metabolism , Nucleoside Diphosphate Sugars/metabolism , Animals , Chick Embryo , Guanosine Diphosphate Sugars/pharmacology , Hydroxylation , Kinetics , Mannosephosphates/pharmacology , Mannosyltransferases/antagonists & inhibitors , Microsomes/enzymology , Structure-Activity Relationship , Substrate Specificity
4.
Biochem J ; 253(3): 795-800, 1988 Aug 01.
Article in English | MEDLINE | ID: mdl-2460084

ABSTRACT

A particulate enzyme preparation made from suspension-cultured dwarf-French-bean (Phaseolus vulgaris) cv. Canadian Wonder cells was shown to incorporate xylose from UDP-D-[14C]xylose into polysaccharide. The reaction was dependent upon the presence of UDP-D-glucose and was stimulated, and apparently protected, by GDP-D-glucose and GDP-D-mannose, though neither was able to replace UDP-D-glucose as a glycosyl donor. The product of the reaction was identified as xyloglucan by analysis of products of enzyme breakdown and acid hydrolysis. Mr determination after proteinase K digestion indicated that the nascent xyloglucan is closely associated with protein. Preincubation of the enzyme with UDP-D-glucose stimulated incorporation from UDP-D-[14C]xylose, suggesting an 'imprecise' mechanism of biosynthesis, as defined by Waldron & Brett [(1985) in Biochemistry of Plant Cell Walls (Brett, C. T. & Hillman, J. R., eds.) (SEB Semin. Ser. 28), pp. 79-97, Cambridge University Press, Cambridge].


Subject(s)
Glucans , Glucosyltransferases/metabolism , Pentosyltransferases/metabolism , Polysaccharides/biosynthesis , Xylans , Cations, Divalent/pharmacology , Chromatography, Gel , Fabaceae/drug effects , Fabaceae/enzymology , Guanosine Diphosphate Mannose/pharmacology , Guanosine Diphosphate Sugars/pharmacology , Plants, Medicinal , Protein Binding , Uridine Diphosphate Glucose/pharmacology , Uridine Diphosphate Xylose/pharmacology , UDP Xylose-Protein Xylosyltransferase
6.
Biochim Biophys Acta ; 570(2): 271-83, 1979 Oct 11.
Article in English | MEDLINE | ID: mdl-40602

ABSTRACT

A 23 000-fold purification of porcine fucokinase (ATP:6-deoxy-L-galactose 1-phosphotransferase, EC 2.7.1.52) has been achieved using a combination of ion-exchange, hydrophobic ligand, affinity, hydroxyapatite and molecular sieve chromatography. The enzyme was determined to have a subunit molecular weight of 78 180 +/- 4260 by sodium dodecyl sulfate chromatography and a tetrameric molecular weight of 309 200 +/- 4100 in the active state as determined by molecular sieve chromatography. The enzyme exhibits a single pH optimum at a pH value of 6.5 and gives evidence of a high order of specificity for L-fucose and ATP. The enzyme requires a divalent metal ion and this need is best satisfied by Mg2+. The activity of the enzyme is modified by a number of nucleotides. ADP is an enzyme inhibitor competitive with ATP. GDP-beta-L-fucose is also an inhibitor and appears to compete with L-fucose. GDP-alpha-D-mannose stimulates the enzyme. A possible role for the actions of these nucleotide sugars is discussed.


Subject(s)
Phosphotransferases/isolation & purification , Thyroid Gland/enzymology , Animals , Fucose , Guanosine Diphosphate Sugars/pharmacology , Hydrogen-Ion Concentration , Kinetics , Methods , Molecular Weight , Nucleotides/pharmacology , Phosphotransferases (Alcohol Group Acceptor) , Substrate Specificity , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...