Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Scand Cardiovasc J ; 53(6): 355-360, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31476919

ABSTRACT

Objectives: Protamine reduces platelet aggregation after cardiopulmonary bypass (CPB). We studied the inhibitory effect of a reduced protamine dose, the duration of impaired platelet function and the possible correlation to postoperative bleeding. Design: Platelet function was assessed by impedance aggregometry in 30 patients undergoing cardiac surgery with CPB at baseline, before protamine administration, after 70% and 100% of the calculated protamine dose, after 20 minutes and at arrival to the intensive care unit. Adenosine diphosphate (ADP), thrombin receptor activating peptide-6 (TRAP), arachidonic acid (AA) and collagen (COL) were used as activators. Blood loss was measured during operation and three hours after surgery. Results are presented as median (25th-75th percentile). Results: Platelet aggregation decreased markedly after the initial dose of protamine (70%) with all activators; ADP 89 (71-110) to 54 (35-78), TRAP 143 (116-167) to 109 (77-136), both p < .01; AA 25 (16-49) to 17 (12-24) and COL 92 (47-103) to 60 (38-81) U, both p < .05. No further decrease was seen after 100% protamine. The effect was transient and after twenty minutes platelet aggregation had started to recover; ADP 76 (54-106), TRAP 138 (95-158), AA 20 (10-35), COL 70 (51-93) U. Blood loss during operation correlated to aggregometry measured at baseline and after protaminization. Conclusions: Protamine after CPB induces a marked decrease in platelet aggregation already at a protamine-heparin ratio of 0.7:1. The impairment seems to be transient and recovery had started after 20 minutes.


Subject(s)
Coronary Artery Bypass , Heart Valve Prosthesis Implantation , Heparin Antagonists/adverse effects , Platelet Aggregation/drug effects , Protamines/adverse effects , Aged , Blood Loss, Surgical/prevention & control , Cardiopulmonary Bypass , Coronary Artery Bypass/adverse effects , Dose-Response Relationship, Drug , Erythrocyte Transfusion , Female , Heart Valve Prosthesis Implantation/adverse effects , Heparin Antagonists/administration & dosage , Humans , Male , Middle Aged , Platelet Function Tests , Postoperative Hemorrhage/etiology , Postoperative Hemorrhage/therapy , Protamines/administration & dosage , Time Factors , Treatment Outcome
2.
New Microbes New Infect ; 15: 6-8, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27830080

ABSTRACT

Catabacter hongkongensis was isolated and cultured from human blood for the first time in Scandinavia. The patient, an 83-year-old man from Dalarna, Sweden, recovered without antibiotic treatment, although a high mortality rate associated with C. hongkongensis infection had been reported from China, Canada and France. The genome of the strain ABBA15k was sequenced, assembled and analysed. In contrast to the type strain of the species HKU16T, no antibiotic resistance was observed in Scandinavian strain ABBA15k. The strain was deposited as CCUG 68271, and the draft genome sequence is available from the DNA Data Bank of Japan (DDBJ), the European Molecular Biology Laboratory (EMBL), and GenBank under the accession number LLYX00000000.

3.
Plant Mol Biol ; 46(3): 335-46, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11488480

ABSTRACT

Two S-adenosylmethionine synthetase (SAMS) cDNAs, PcSAMS1 and PcSAMS2, have been identified in Pinus contorta. We found that the two genes are differentially expressed during root development. Thus, PcSAMS1 is preferentially expressed in roots and exhibits a specific expression pattern in the meristem at the onset of adventitious root development, whereas PcSAMS2 is expressed in roots as well as in shoots and is down-regulated during adventitious root formation. The expression of the two SAMS genes is different from the SAMS activity levels during adventitious root formation. We conclude that other SAMS genes that remain to be characterized may contribute to the observed SAMS activity, or that the activities of PcSAMS1 and PcSAMS2 are affected by post-transcriptional regulation. The deduced amino acid sequences of PcSAMS1 and PcSAMS2 are highly divergent, suggesting different functional roles. However, both carry the two perfectly conserved motifs that are common to all plant SAMS. At the protein level, PcSAMS2 shares about 90% identity to other isolated eukaryotic SAMS, while PcSAMS1 shares less than 50% identity with other plant SAMS. In a phylogenetic comparison, PcSAMS1 seems to have diverged significantly from all other SAMS genes. Nevertheless, PcSAMS1 was able to complement a Saccharomyces cerevisiae sam1 sam2 double mutant, indicating that it encodes a functional SAMS enzyme.


Subject(s)
Cycadopsida/genetics , Methionine Adenosyltransferase/genetics , Plant Roots/genetics , Amino Acid Sequence , Cycadopsida/enzymology , Cycadopsida/growth & development , DNA, Complementary/chemistry , DNA, Complementary/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Plant , Genetic Complementation Test , Hypocotyl/drug effects , Hypocotyl/enzymology , Hypocotyl/genetics , In Situ Hybridization , Indoleacetic Acids/pharmacology , Indoles/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Methionine Adenosyltransferase/metabolism , Molecular Sequence Data , Mutation , Phylogeny , Plant Roots/drug effects , Plant Roots/growth & development , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution , Transcription, Genetic
4.
Science ; 288(5469): 1248-51, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10818001

ABSTRACT

A high proportion of purebred Hampshire pigs carries the dominant RN- mutation, which causes high glycogen content in skeletal muscle. The mutation has beneficial effects on meat content but detrimental effects on processing yield. Here, it is shown that the mutation is a nonconservative substitution (R200Q) in the PRKAG3 gene, which encodes a muscle-specific isoform of the regulatory gamma subunit of adenosine monophosphate-activated protein kinase (AMPK). Loss-of-function mutations in the homologous gene in yeast (SNF4) cause defects in glucose metabolism, including glycogen storage. Further analysis of the PRKAG3 signaling pathway may provide insights into muscle physiology as well as the pathogenesis of noninsulin-dependent diabetes mellitus in humans, a metabolic disorder associated with impaired glycogen synthesis.


Subject(s)
Glycogen/metabolism , Muscle, Skeletal/enzymology , Point Mutation , Protein Kinases/genetics , AMP-Activated Protein Kinases , Alleles , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Blotting, Northern , Cloning, Molecular , DNA, Complementary/isolation & purification , Gene Expression Regulation, Enzymologic , Homozygote , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/isolation & purification , Molecular Sequence Data , Muscle, Skeletal/metabolism , Organ Specificity/genetics , Phenotype , Protein Kinases/biosynthesis , Protein Kinases/isolation & purification , Sequence Homology, Amino Acid , Swine
5.
J Mol Biol ; 262(5): 706-20, 1996 Oct 11.
Article in English | MEDLINE | ID: mdl-8876648

ABSTRACT

The three-dimensional structure of mouse ribonucleotide reductase R2 has been determined at 2.3 A resolution using molecular replacement and refined to an R-value of 19.1% (Rfree = 25%) with good stereo-chemistry. The overall tertiary structure architecture of mouse R2 is similar to that from Escherichia coli R2. However, several important structural differences are observed. Unlike the E. coli protein, the mouse dimer is completely devoid of beta-strands. The sequences differ significantly between the mouse and E. coli R2s, but there is high sequence identity among the eukaryotic R2 proteins, and the identities are localized over the whole sequence. Therefore, the three-dimensional structures of other mammalian ribonucleotide reductase R2 proteins are expected to be very similar to that of the mouse enzyme. In mouse R2 a narrow hydrophobic channel leads to the proposed binding site for molecular oxygen near to the iron-radical site in the interior of the protein. In E. coli R2 this channel is blocked by the phenyl ring of a tyrosine residue, which in mouse R2 is a serine. These structural variations may explain the observed differences in sensitivity to radical scavengers. The structure determination is based on diffraction data from crystals grown at pH 4.7. Unexpectedly, the protein is not iron-free, but contains one iron ion bound at one of the dinuclear iron sites. This ferric ion is bound with partial occupancy and is coordinated by three glutamic acids (one bidentate) and one histidine in a bipyramidal coordination that has a free apical coordination position. Soaking of crystals in a solution of ferrous salt at pH 4.7 increased the occupancy on the already occupied site, but without any detectable binding at the second site.


Subject(s)
Bacterial Proteins/ultrastructure , Ribonucleotide Reductases/ultrastructure , Animals , Crystallography, X-Ray , Escherichia coli/chemistry , Free Radical Scavengers/chemistry , Hydroxyurea/chemistry , Iron/chemistry , Mice , Models, Molecular , Oxidation-Reduction , Protein Conformation , Protein Structure, Secondary , Ribonucleotide Reductases/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Tyrosine/chemistry
6.
FEBS Lett ; 373(3): 310-2, 1995 Oct 16.
Article in English | MEDLINE | ID: mdl-7589490

ABSTRACT

The R2 protein component of mouse ribonucleotide reductase has been obtained from overproducing Escherichia coli bacteria. It has been crystallized using NaCl as precipitant. The crystals are orthorhombic, space group C222(1), with cell dimensions a = 76.9 A, b = 108.9 A, c = 92.7 A and diffract to at least 2.5 A. The asymmetric unit of the crystals contains one monomer. Rotation and translation function searches using a model based on the weakly homologous E. coli R2 gave one significant peak. Rotation about a crystallographic 2-fold axis parallel to the a-axis produces an R2 dimer with dimer interactions very similar to those found for E. coli R2.


Subject(s)
Ribonucleotide Reductases/chemistry , Animals , Crystallization , Crystallography, X-Ray , Fourier Analysis , Iron/analysis , Mice , Protein Conformation , Recombinant Proteins/chemistry
7.
J Biol Chem ; 269(37): 23171-6, 1994 Sep 16.
Article in English | MEDLINE | ID: mdl-8083221

ABSTRACT

Mammalian ribonucleotide reductase is a heterotetramer formed by the two non-identical homodimers proteins R1 and R2. We have succeeded in expressing the 90-kDa mouse R1 protein in Escherichia coli in an active, soluble form using the T7 RNA polymerase pET vector system. To avoid inclusion bodies, the bacteria were grown at 15 degrees C with minimal concentration of the inducer isopropyl-1-thio-beta-D-galactopyranoside. After a rapid purification procedure, approximately 20 mg of pure R1 protein were obtained per liter of bacterial culture. The concentrated R1 protein solution had a pinkish red color. Spectroscopy in combination with iron and labile sulfur analyses demonstrated that the color originated from an iron-sulfur complex. However, all attempts to demonstrate a function of this complex have been inconclusive. A comparison of the recombinant R1 protein with the corresponding protein purified from calf thymus showed no evidence for glycosylation. Circular dichroism spectroscopy indicated an alpha-helical content of 50%. A flexible COOH-terminal tail of 7 residues in the R2 protein was earlier shown to be essential for binding to the R1 protein. Using a peptide protection assay and photoaffinity labeling, we now show that the R2 protein tail interacts with a region close to the carboxyl terminus of the R1 protein.


Subject(s)
Ribonucleotide Reductases/isolation & purification , Affinity Labels , Amino Acid Sequence , Animals , Base Sequence , Carbohydrates/analysis , Cattle , Circular Dichroism , Electron Spin Resonance Spectroscopy , Electrophoresis, Polyacrylamide Gel , Iron , Mice , Molecular Sequence Data , Peptides , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ribonucleotide Reductases/metabolism , Sulfur
8.
EMBO J ; 8(9): 2475-9, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2684652

ABSTRACT

Mammalian ribonucleotide reductase consists of two non-identical subunits, proteins M1 and M2. M2-related DNA sequences are present on mouse chromosomes 4, 7, 12 and 13. However, M2-overproducing mouse cells show amplification of a chromosome 12-specific, single 13 kb HindIII fragment, which probably represents the active gene. We have isolated this fragment from parental mouse cell DNA and used it to clone and characterize the functional M2 gene. The 5770 bp transcribed M2 sequence contains ten exons separated by nine 95-917 bp introns. The 501 bp of 5' flanking DNA is G + C rich and contains TTTAAA and CCAAT sequences as well as potential Sp1 binding sites. The M2-related sequence on chromosome 13, which contains only the last six exons and several internal rearrangements, is a pseudogene. Transfection of BALB/3T3 cells with the M2 gene resulted in stable transformants with a 10-fold reduction in sensitivity to hydroxyurea, compared to control cells. This confirmed that the cloned M2 genomic DNA represents the functional gene and conclusively establishes the link between hydroxyurea resistance and M2 expression in mammalian cells. M2 genomic DNA should be a valuable dominant, selectable marker for identifying and isolating stable co-transformants.


Subject(s)
Ribonucleotide Reductases/genetics , Animals , Base Sequence , Carbocyanines/metabolism , Cells, Cultured , Cloning, Molecular , Drug Resistance/genetics , Exons , Genetic Markers , Hydroxyurea/pharmacology , Introns , Lypressin/metabolism , Lypressin/pharmacology , Mice , Molecular Sequence Data , Pseudogenes/genetics , Rhodamines/metabolism , Rhodamines/pharmacology , Transcription, Genetic , Transfection
9.
J Biol Chem ; 260(5): 2737-41, 1985 Mar 10.
Article in English | MEDLINE | ID: mdl-3882700

ABSTRACT

The M2 subunit of mammalian ribonucleotide reductase was purified to homogeneity from hydroxyurea-resistant, M2-overproducing mouse cells. The purification procedure involved affinity chromatography on an anti-tubulin antibody-Sepharose column and high performance gel permeation chromatography. The pure protein is a dimer of Mr = 88,000, containing stoichiometric amounts of a non-heme iron center and a tyrosyl free radical. The radical is destroyed by hydroxyurea but can readily be regenerated on incubation of the radical-free protein alone with iron-dithiothreitol in the presence of air. The ability to spontaneously regenerate the tyrosyl radical distinguishes protein M2 from the corresponding subunit of Escherichia coli ribonucleotide reductase, protein B2, but apart from that the two proteins are very similar.


Subject(s)
Ribonucleotide Reductases/analysis , Animals , Antibodies, Monoclonal , Chromatography, Affinity , Chromatography, DEAE-Cellulose , Electrophoresis, Polyacrylamide Gel , Escherichia coli/enzymology , Macromolecular Substances , Mice , Mice, Inbred A , Molecular Weight , Spectrophotometry , Tubulin/immunology
10.
Biochem Biophys Res Commun ; 110(3): 859-65, 1983 Feb 10.
Article in English | MEDLINE | ID: mdl-6340669

ABSTRACT

A radical-free preparation of a highly purified ribonucleotide reductase from calf thymus was shown to generate an M2-specific tyrosine free radical on incubation with iron and dithiothreitol in the presence of air. The radical is essential for activity but once formed has a half-life of about 10 min. Using the calf thymus enzyme, there is a continual requirement of oxygen and iron for ribonucleotide reduction indicating a continual regeneration of the radical during enzyme catalysis. We therefore propose that one way a cell may regulate ribonucleotide reductase activity is by controlling the generation of M2-specific tyrosine free radicals within existing M2 molecules.


Subject(s)
Iron/pharmacology , Ribonucleotide Reductases/metabolism , Aerobiosis , Anaerobiosis , Animals , Cattle , Dithiothreitol/pharmacology , Free Radicals , Kinetics , Oxygen , Thymus Gland/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...