Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Biol ; 22(12): 1095-101, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22608508

ABSTRACT

Life cycle adaptation to latitudinal and seasonal variation in photoperiod and temperature is a major determinant of evolutionary success in flowering plants. Whereas the life cycle of the dicotyledonous model species Arabidopsis thaliana is controlled by two epistatic genes, FLOWERING LOCUS C and FRIGIDA, three unrelated loci (VERNALIZATION) determine the spring and winter habits of monocotyledonous plants such as temperate cereals. In the core eudicot species Beta vulgaris, whose lineage diverged from that leading to Arabidopsis shortly after the monocot-dicot split 140 million years ago, the bolting locus B is a master switch distinguishing annuals from biennials. Here, we isolated B and show that the pseudo-response regulator gene BOLTING TIME CONTROL 1 (BvBTC1), through regulation of the FLOWERING LOCUS T genes, is absolutely necessary for flowering and mediates the response to both long days and vernalization. Our results suggest that domestication of beets involved the selection of a rare partial loss-of-function BvBTC1 allele that imparts reduced sensitivity to photoperiod that is restored by vernalization, thus conferring bienniality, and illustrate how evolutionary plasticity at a key regulatory point can enable new life cycle strategies.


Subject(s)
Adaptation, Biological/physiology , Agriculture/methods , Beta vulgaris/physiology , Biological Evolution , Flowers/physiology , Genes, Regulator/genetics , Plant Proteins/genetics , Adaptation, Biological/genetics , Amino Acid Sequence , Amplified Fragment Length Polymorphism Analysis , Base Sequence , Beta vulgaris/genetics , Chromosome Mapping , Chromosomes, Artificial, Bacterial/genetics , Cloning, Molecular , DNA Primers/genetics , Flowers/genetics , Genetic Markers/genetics , Haplotypes/genetics , Immunoblotting , Models, Biological , Molecular Sequence Data , Phenotype , Photoperiod , Phylogeny , Seasons , Selection, Genetic , Sequence Alignment , Sequence Analysis, DNA
2.
Biochem J ; 433(2): 383-91, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21029045

ABSTRACT

NOSs (nitric oxide synthases) catalyse the oxidation of L-arginine to L-citrulline and nitric oxide via the intermediate NOHA (N(ω)-hydroxy-L-arginine). This intermediate is rapidly converted further, but to a small extent can also be liberated from the active site of NOSs and act as a transportable precursor of nitric oxide or potent physiological inhibitor of arginases. Thus its formation is of enormous importance for the nitric-oxide-generating system. It has also been shown that NOHA is reduced by microsomes and mitochondria to L-arginine. In the present study, we show for the first time that both human isoforms of the newly identified mARC (mitochondrial amidoxime reducing component) enhance the rate of reduction of NOHA, in the presence of NADH cytochrome b5 reductase and cytochrome b5, by more than 500-fold. Consequently, these results provide the first hints that mARC might be involved in mitochondrial NOHA reduction and could be of physiological significance in affecting endogenous nitric oxide levels. Possibly, this reduction represents another regulative mechanism in the complex regulation of nitric oxide biosynthesis, considering a mitochondrial NOS has been identified. Moreover, this reduction is not restricted to NOHA since the analogous arginase inhibitor NHAM (N(ω)-hydroxy-N(δ)-methyl-L-arginine) is also reduced by this system.


Subject(s)
Arginine/analogs & derivatives , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Animals , Arginine/metabolism , Benzamidines , Hep G2 Cells , Humans , Oxidation-Reduction , Swine
3.
J Biol Chem ; 285(48): 37847-59, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20861021

ABSTRACT

The mitochondrial amidoxime reducing component mARC is a newly discovered molybdenum enzyme that is presumed to form the catalytical part of a three-component enzyme system, consisting of mARC, heme/cytochrome b(5), and NADH/FAD-dependent cytochrome b(5) reductase. mARC proteins share a significant degree of homology to the molybdenum cofactor-binding domain of eukaryotic molybdenum cofactor sulfurase proteins, the latter catalyzing the post-translational activation of aldehyde oxidase and xanthine oxidoreductase. The human genome harbors two mARC genes, referred to as hmARC-1/MOSC-1 and hmARC-2/MOSC-2, which are organized in a tandem arrangement on chromosome 1. Recombinant expression of hmARC-1 and hmARC-2 proteins in Escherichia coli reveals that both proteins are monomeric in their active forms, which is in contrast to all other eukaryotic molybdenum enzymes that act as homo- or heterodimers. Both hmARC-1 and hmARC-2 catalyze the N-reduction of a variety of N-hydroxylated substrates such as N-hydroxy-cytosine, albeit with different specificities. Reconstitution of active molybdenum cofactor onto recombinant hmARC-1 and hmARC-2 proteins in the absence of sulfur indicates that mARC proteins do not belong to the xanthine oxidase family of molybdenum enzymes. Moreover, they also appear to be different from the sulfite oxidase family, because no cysteine residue could be identified as a putative ligand of the molybdenum atom. This suggests that the hmARC proteins and sulfurase represent members of a new family of molybdenum enzymes.


Subject(s)
Mitochondria/enzymology , Mitochondrial Proteins/chemistry , Molybdenum/metabolism , Multigene Family , Oxidoreductases/chemistry , Animals , Humans , Kinetics , Mice , Mitochondria/chemistry , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Spectrum Analysis , Substrate Specificity , Sulfite Oxidase/chemistry , Sulfite Oxidase/genetics , Sulfite Oxidase/metabolism
4.
Drug Metab Dispos ; 38(11): 1917-21, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20699408

ABSTRACT

Purification of the mitochondrial enzyme responsible for reduction of N-hydroxylated amidine prodrugs led to the identification of two newly discovered mammalian molybdenum-containing proteins, the mitochondrial amidoxime reducing components mARC-1 and mARC-2 (Gruenewald et al., 2008). These 35-kDa proteins represent a novel group of molybdenum proteins in eukaryotes as they form a molybdenum cofactor-dependent enzyme system consisting of three separate proteins (Havemeyer et al., 2006). Each mARC protein reduces N-hydroxylated compounds after reconstitution with the electron transport proteins cytochrome b(5) and b(5) reductase. In continuation of our drug metabolism investigations (Havemeyer et al., 2006; Gruenewald et al., 2008), we present data from reconstituted enzyme systems with recombinant human and native porcine enzymes showing the reduction of N-hydroxy-sulfonamides (sulfohydroxamic acids) to sulfonamides: the N-hydroxy-sulfonamide N-hydroxy-valdecoxib (N-hydroxy-4-[5-methyl-3-phenyl-4-isoxazolyl]-benzenesulfonamide) represents a novel cyclooxygenase (COX)-2 inhibitor and is therefore a drug candidate in the treatment of diseases associated with rheumatic inflammation, pain, and fever. It was synthesized as an analog of the known COX-2 inhibitor valdecoxib (4-[5-methyl-3-phenyl-4-isoxazolyl]-benzenesulfonamide) (Talley et al., 2000). N-Hydroxy-valdecoxib had low in vitro COX-2 activity but showed significant analgesic activity in vivo and a prolonged therapeutic effect compared with valdecoxib (Erdélyi et al., 2008). In this report, we demonstrate that N-hydroxy-valdecoxib is enzymatically reduced to its pharmacologically active metabolite valdecoxib. Thus, N-hydroxy-valdecoxib acts as prodrug that is activated by the molybdenum-containing enzyme mARC.


Subject(s)
Isoxazoles/metabolism , Metalloproteins , Mitochondrial Proteins/metabolism , Molybdenum , Oxidoreductases/metabolism , Prodrugs/metabolism , Sulfonamides/metabolism , Animals , Chromatography, High Pressure Liquid , Cytochrome-B(5) Reductase/genetics , Cytochrome-B(5) Reductase/metabolism , Cytochromes b5/genetics , Cytochromes b5/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , In Vitro Techniques , Isoxazoles/chemistry , Metabolic Detoxication, Phase I , Metalloproteins/genetics , Metalloproteins/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Molecular Structure , Molybdenum/metabolism , Oxidation-Reduction , Oxidoreductases/genetics , Prodrugs/chemistry , Substrate Specificity , Sulfonamides/chemistry , Swine , Transfection
5.
J Med Chem ; 51(24): 8173-7, 2008 Dec 25.
Article in English | MEDLINE | ID: mdl-19053771

ABSTRACT

The recently discovered mammalian molybdoprotein mARC1 is capable of reducing N-hydroxylated compounds. Upon reconstitution with cytochrome b(5) and b(5) reductase, benzamidoxime, pentamidine, and diminazene amidoximes, N-hydroxymelagatran, guanoxabenz, and N-hydroxydebrisoquine are efficiently reduced. These substances are amidoxime/N-hydroxyguanidine prodrugs, leading to improved bioavailability compared to the active amidines/guanidines. Thus, the recombinant enzyme allows prediction about in vivo reduction of N-hydroxylated prodrugs. Furthermore, the prodrug principle is not dependent on cytochrome P450 enzymes.


Subject(s)
Molybdenum/chemistry , Oxidoreductases/chemistry , Prodrugs/chemistry , Animals , Benzamidines/chemistry , Cattle , Cell Line , Chemistry, Pharmaceutical/methods , Cloning, Molecular , Cytochrome-B(5) Reductase/chemistry , Drug Design , Humans , Ligands , Models, Chemical , Oxidoreductases/metabolism , Recombinant Proteins/chemistry , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...