Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nucleosides Nucleotides Nucleic Acids ; 39(10-12): 1281-1305, 2020.
Article in English | MEDLINE | ID: mdl-32043431

ABSTRACT

This paper is based on the Anne Simmonds Memorial Lecture, given by Monika Löffler at the International Symposium on Purine and Pyrimidine Metabolism in Man, Lyon 2019. It is dedicated to H. Anne Simmonds (died 2010) - a founding member of the ESSPPMM, since 2003 Purine and Pyrimidine Society - and her outstanding contributions to the identification and study of inborn errors of purine and pyrimidine metabolism. The distinctive intracellular arrangement of pyrimidine de novo synthesis in higher eukaryotes is important to cells with a high demand for nucleic acid synthesis. The proximity of the enzyme active sites and the resulting channeling in CAD and UMP synthase is of kinetic benefit. The intervening enzyme dihydroorotate dehydrogenase (DHODH) is located in the mitochondrion with access to the ubiquinone pool, thus ensuring efficient removal of redox equivalents through the constitutive activity of the respiratory chain, also a mechanism through which the input of 2 ATP for carbamylphosphate synthesis is balanced by Oxphos. The obligatory contribution of O2 to de novo UMP synthesis means that DHODH has a pivotal role in adapting the proliferative capacity of cells to different conditions of oxygenation, such as hypoxia in growing tumors. DHODH also is a validated drug target in inflammatory diseases. This survey of selected topics of personal interest and reflection spans some 40 years of our studies from tumor cell cultures under hypoxia to in vitro assays including purification from mitochondria, localization, cloning, expression, biochemical characterization, crystallisation, kinetics and inhibition patterns of eukaryotic DHODH enzymes.


Subject(s)
Mitochondria/enzymology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pyrimidines/metabolism , Dihydroorotate Dehydrogenase , Electron Transport , Humans , Mitochondria/metabolism
2.
Front Microbiol ; 10: 1479, 2019.
Article in English | MEDLINE | ID: mdl-31316493

ABSTRACT

The oomycete Phytophthora infestans is the causal agent of tomato and potato late blight, a disease that causes tremendous economic losses in the production of solanaceous crops. The similarities between oomycetes and the apicomplexa led us to hypothesize that dihydroorotate dehydrogenase (DHODH), the enzyme catalyzing the fourth step in pyrimidine biosynthetic pathway, and a validated drug target in treatment of malaria, could be a potential target for controlling P. infestans growth. In eukaryotes, class 2 DHODHs are mitochondrially associated ubiquinone-linked enzymes that catalyze the fourth, and only redox step of de novo pyrimidine biosynthesis. We characterized the enzymes from both the pathogen and a host, Solanum tuberosum. Plant DHODHs are known to be class 2 enzymes. Sequence analysis suggested that the pathogen enzyme (PiDHODHs) also belongs to this class. We confirmed the mitochondrial localization of GFP-PiDHODH showing colocalization with mCherry-labeled ATPase in a transgenic pathogen. N-terminally truncated versions of the two DHODHs were overproduced in E. coli, purified, and kinetically characterized. StDHODH exhibited a apparent specific activity of 41 ± 1 µmol min-1 mg-1, a kcat app of 30 ± 1 s-1, and a Km app of 20 ± 1 µM for L-dihydroorotate, and a Km app= 30 ± 3 µM for decylubiquinone (Qd). PiDHODH exhibited an apparent specific activity of 104 ± 1 µmol min-1 mg-1, a kcat app of 75 ± 1 s-1, and a Km app of 57 ± 3 µM for L-dihydroorotate, and a Km app of 15 ± 1 µM for Qd. The two enzymes exhibited different activities with different quinones and napthoquinone derivatives, and different sensitivities to compounds known to cause inhibition of DHODHs from other organisms. The IC50 for A77 1726, a nanomolar inhibitor of human DHODH, was 2.9 ± 0.6 mM for StDHODH, and 79 ± 1 µM for PiDHODH. In vivo, 0.5 mM A77 1726 decreased mycelial growth by approximately 50%, after 92 h. Collectively, our findings suggest that the PiDHODH could be a target for selective inhibitors and we provide a biochemical background for the development of compounds that could be helpful for the control of the pathogen, opening the way to protein crystallization.

3.
J Genet Genomics ; 42(5): 207-19, 2015 May 20.
Article in English | MEDLINE | ID: mdl-26059769

ABSTRACT

It is timely to consider the many facets of the small molecule orotic acid (OA), which is well-known as an essential intermediate of pyrimidine de novo synthesis. In addition, it can be taken up by erythrocytes and hepatocytes for conversion into uridine and for use in the pyrimidine recycling pathway. We discuss the link between dietary orotate and fatty liver in rats, and the potential for the alleviation of neonatal hyperbilirubinaemia. We address the development of orotate derivatives for application as anti-pyrimidine drugs, and of complexes with metal ions and organic cations to assist therapies of metabolic syndromes. Recent genetic data link human Miller syndrome to defects in the dihydroorotate dehydrogenase (DHODH) gene, hence to depleted orotate production. Another defect in pyrimidine biosynthesis, the orotic aciduria arising in humans and cattle with a deficiency of UMP synthase (UMPS), has different symptoms. More recent work leads us to conclude that OA may have a role in regulating gene transcription.


Subject(s)
Orotic Acid/metabolism , Pyrimidines/biosynthesis , Animals , Central Nervous System/metabolism , Enzymes/metabolism , Humans , Milk/metabolism
4.
Mol Biochem Parasitol ; 184(2): 71-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22580100

ABSTRACT

The pyrimidine biosynthesis pathway in the protozoan pathogen Toxoplasma gondii is essential for parasite growth during infection. To investigate the properties of dihydroorotate dehydrogenase (TgDHOD), the fourth enzyme in the T. gondii pyrimidine pathway, we expressed and purified recombinant TgDHOD. TgDHOD exhibited a specific activity of 84U/mg, a k(cat) of 89s(-1), a K(m)=60µM for l-dihydroorotate, and a K(m)=29µM for decylubiquinone (Q(D)). Quinones lacking or having short isoprenoid side chains yielded lower k(cat)s than Q(D). As expected, fumarate was a poor electron acceptor for this family 2 DHOD. The IC(50)s determined for A77-1726, the active derivative of the human DHOD inhibitor leflunomide, and related compounds MD249 and MD209 were, 91µM, 96µM, and 60µM, respectively. The enzyme was not significantly affected by brequinar or TTFA, known inhibitors of human DHOD, or by atovaquone. DSM190, a known inhibitor of Plasmodium falciparum DHOD, was a poor inhibitor of TgDHOD. TgDHOD exhibits a lengthy 157-residue N-terminal extension, consistent with a potential organellar targeting signal. We constructed C-terminally c-myc tagged TgDHODs to examine subcellular localization of TgDHOD in transgenic parasites expressing the tagged protein. Using both exogenous and endogenous expression strategies, anti-myc fluorescence signal colocalized with antibodies against the mitochondrial marker ATPase. These findings demonstrate that TgDHOD is associated with the parasite's mitochondrion, revealing this organelle as the site of orotate production in T. gondii. The TgDHOD gene appears to be essential because while gene tagging was successful at the TgDHOD gene locus, attempts to delete the TgDHOD gene were not successful in the KU80 background. Collectively, our study suggests that TgDHOD is an excellent target for the development of anti-Toxoplasma drugs.


Subject(s)
Mitochondria/enzymology , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Protozoan Proteins/chemistry , Pyrimidines/biosynthesis , Toxoplasma/enzymology , Amino Acid Sequence , Biosynthetic Pathways , Cloning, Molecular , Conserved Sequence , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemistry , Gene Knockout Techniques , Kinetics , Molecular Sequence Data , Orotic Acid/analogs & derivatives , Orotic Acid/chemistry , Oxidation-Reduction , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Protein Sorting Signals , Protein Transport , Proteolysis , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
5.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1147-54, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22132969

ABSTRACT

Tissue-specific expression of the genes coding for the six enzymes of the de novo pyrimidine synthesis and for the first enzyme of the degradation pathway, dihydropyrimidine dehydrogenase (DPD), was analyzed in the rat using the in situ hybridization technique. Transcripts of the biosynthetic enzymes were detected in liver, kidney, and spleen with the highest expression in the white pulp. DPD was also transcribed in these organs with a striking layer-specific localization of DPD mRNA and protein in the kidney. All enzyme mRNAs were present in brain at low levels, but with region- and cell-specific differences. The relatively high expression in cortical regions including cerebellum and hippocampus points to a fundamental role of pyrimidine metabolism in brain function.


Subject(s)
Brain/enzymology , Gene Expression Regulation, Enzymologic , Organ Specificity , Pyrimidines/biosynthesis , Animals , Dihydrouracil Dehydrogenase (NADP)/genetics , Dihydrouracil Dehydrogenase (NADP)/metabolism , Immunohistochemistry , Kidney/cytology , Kidney/enzymology , Male , Rats , Rats, Wistar
6.
FEMS Yeast Res ; 7(6): 897-904, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17617217

ABSTRACT

In all organisms the fourth catalytic step of the pyrimidine biosynthesis is driven by the flavoenzyme dihydroorotate dehydrogenase (DHODH, EC 1.3.99.11). Cytosolic DHODH of the established model organism Saccharomyces cerevisiae catalyses the oxidation of dihydroorotate to orotate and the reduction of fumarate to succinate. Here, we investigate the structure and mechanism of DHODH from S. cerevisiae and show that the recombinant ScDHODH exists as a homodimeric enzyme in vitro. Inhibition of ScDHODH by the reaction product was observed and kinetic studies disclosed affinity for orotate (K(ic)=7.7 microM; K(ic) is the competitive inhibition constant). The binding constant for orotate was measured through comparison of UV-visible spectra of the bound and unbound recombinant enzyme. The midpoint reduction potential of DHODH-bound flavine mononucleotide determined from analysis of spectral changes was -242 mV (vs. NHE) under anaerobic conditions. A search for alternative electron acceptors revealed that homologues such as mesaconate can be used as electron acceptors.


Subject(s)
Fumarates/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Catalysis , Dihydroorotate Dehydrogenase , Metabolic Networks and Pathways , Models, Molecular , Oxidation-Reduction , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/isolation & purification , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spectrum Analysis
7.
Appl Environ Microbiol ; 73(10): 3371-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17369345

ABSTRACT

Dihydroorotate dehydrogenase (DHODH; EC 1.3.99.11) is a central enzyme of pyrimidine biosynthesis and catalyzes the oxidation of dihydroorotate to orotate. DHODH is an important target for antiparasitic and cytostatic drugs since rapid cell proliferation often depends on the de novo synthesis of pyrimidine nucleotides. We have cloned the pyr4 gene encoding mitochondrial DHODH from the basidiomycetous plant pathogen Ustilago maydis. We were able to show that pyr4 contains a functional mitochondrial targeting signal. The deletion of pyr4 resulted in uracil auxotrophy, enhanced sensitivity to UV irradiation, and a loss of pathogenicity on corn plants. The biochemical characterization of purified U. maydis DHODH overproduced in Escherichia coli revealed that the U. maydis enzyme uses quinone electron acceptor Q6 and is resistant to several commonly used DHODH inhibitors. Here we show that the expression of the human DHODH gene fused to the U. maydis mitochondrial targeting signal is able to complement the auxotrophic phenotype of pyr4 mutants. While U. maydis wild-type cells were resistant to the DHODH inhibitor brequinar, strains expressing the human DHODH gene became sensitive to this cytostatic drug. Such engineered U. maydis strains can be used in sensitive in vivo assays for the development of novel drugs specifically targeted at either human or fungal DHODH.


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Gene Deletion , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/genetics , Ustilago/drug effects , Ustilago/genetics , Biphenyl Compounds/pharmacology , Cloning, Molecular , DNA, Fungal/chemistry , DNA, Fungal/genetics , Dihydroorotate Dehydrogenase , Gene Expression , Genetic Complementation Test , Humans , Mitochondria/enzymology , Molecular Sequence Data , Protein Sorting Signals/genetics , Pyrimidines/biosynthesis , Recombinant Proteins/antagonists & inhibitors , Ustilago/growth & development , Ustilago/metabolism
8.
FEBS J ; 273(14): 3183-91, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16774642

ABSTRACT

Candida albicans is the most prevalent yeast pathogen in humans, and recently it has become increasingly resistant to the current antifungal agents. In this study we investigated C. albicans dihydroorotate dehydrogenase (DHODH, EC 1.3.99.11), which catalyzes the fourth step of de novo pyrimidine synthesis, as a new target for controlling infection. We propose that the enzyme is a member of the DHODH family 2, which comprises mitochondrially bound enzymes, with quinone as the direct electron acceptor and oxygen as the final electron acceptor. Full-length DHODH and N-terminally truncated DHODH, which lacks the targeting sequence and the transmembrane domain, were subcloned from C. albicans, recombinantly expressed in Escherichia coli, purified, and characterized for their kinetics and substrate specificity. An inhibitor screening with 28 selected compounds was performed. Only the dianisidine derivative, redoxal, and the biphenyl quinoline-carboxylic acid derivative, brequinar sodium, which are known to be potent inhibitors of mammalian DHODH, markedly reduced C. albicans DHODH activity. This study provides a background for the development of antipyrimidines with high efficacy for decreasing in situ pyrimidine nucleotide pools in C. albicans.


Subject(s)
Candida albicans/enzymology , Candida albicans/pathogenicity , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Aminobiphenyl Compounds/metabolism , Aminobiphenyl Compounds/pharmacology , Biphenyl Compounds/metabolism , Biphenyl Compounds/pharmacology , Conserved Sequence , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Glutathione Transferase/metabolism , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Kinetics , Molecular Sequence Data , Molecular Structure , Mutation , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
9.
Trends Mol Med ; 11(9): 430-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16098809

ABSTRACT

Genetic defects involving enzymes essential for pyrimidine nucleotide metabolism have provided new insights into the vital physiological functions of these molecules in addition to nucleic acid synthesis. Such aberrations disrupt the haematological, nervous or mitochondrial systems and can cause adverse reactions to analogue therapy. Regulation of pyrimidine pathways is also known to be disrupted in malignancies. Nine genetic defects have now been identified but only one is currently treatable. Diagnosis is aided by the accumulation of specific metabolites. Recently, progress has been made in understanding the molecular mechanisms underlying inborn errors of pyrimidine metabolism, together with the key clinical issues and the implications for the future development of novel drugs and therapeutic strategies.


Subject(s)
Health , Purine-Pyrimidine Metabolism, Inborn Errors/metabolism , Pyrimidines/metabolism , Signal Transduction , Humans , Purine-Pyrimidine Metabolism, Inborn Errors/diagnosis , Purine-Pyrimidine Metabolism, Inborn Errors/genetics , Purine-Pyrimidine Metabolism, Inborn Errors/therapy , Pyrimidines/antagonists & inhibitors
10.
Clin Chem ; 50(11): 2117-24, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15375016

ABSTRACT

BACKGROUND: The concentrations of the pyrimidine "de novo" metabolites and their degradation products in urine are useful indicators for the diagnosis of an inborn error of the pyrimidine de novo pathway or a urea-cycle defect. Until now, no procedure was available that allowed the analysis of all of these metabolites in a single analytical run. We describe a rapid, specific method to measure these metabolites by HPLC-tandem mass spectrometry. METHODS: Urine or urine-soaked filter-paper strips were used to measure N-carbamyl-aspartate, dihydroorotate, orotate, orotidine, uridine, and uracil. Reversed-phase HPLC was combined with electrospray ionization tandem mass spectrometry, and detection was performed by multiple-reaction monitoring. Stable-isotope-labeled reference compounds were used as internal standards. RESULTS: All pyrimidine de novo metabolites and their degradation products were measured within a single analytical run of 14 min with lower limits of detection of 0.4-3 micromol/L. The intra- and interassay variation for urine with added compounds was 1.2-5% for urines and 2-9% for filter-paper extracts of the urines. Recoveries of the added metabolites were 97-106% for urine samples and 97-115% for filter-paper extracts of the urines. Analysis of urine samples from patients with a urea-cycle defect or pyrimidine degradation defect showed an aberrant metabolic profile when compared with controls. CONCLUSION: HPLC with electrospray ionization tandem mass spectrometry allows rapid testing for disorders affecting the pyrimidine de novo pathway. The use of filter-paper strips could facilitate collection, transport, and storage of urine samples.


Subject(s)
Pyrimidines/urine , Specimen Handling/methods , Argininosuccinic Aciduria , Child , Child, Preschool , Chromatography, High Pressure Liquid , Dihydrouracil Dehydrogenase (NADP)/urine , Humans , Infant , Infant, Newborn , Ornithine Carbamoyltransferase Deficiency Disease , Paper , Pyrimidines/metabolism , Reference Values , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization
11.
FEBS Lett ; 568(1-3): 129-34, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15196933

ABSTRACT

Genes for two structurally and functionally different dihydroorotate dehydrogenases (DHODHs, EC 1.3.99.11), catalyzing the fourth step of pyrimidine biosynthesis, have been previously found in yeast Saccharomyces kluyveri. One is closely related to the Schizosaccharomyces pombe mitochondrial family 2 enzymes, which use quinones as direct and oxygen as the final electron acceptor. The other one resembles the Saccharomyces cerevisiae cytosolic family 1A fumarate-utilizing DHODH. The DHODHs from S. kluyveri, Sch. pombe and S. cerevisiae, were expressed in Escherichia coli and compared for their biochemical properties and interaction with inhibitors. Benzoates as pyrimidine ring analogs were shown to be selective inhibitors of cytosolic DHODs. This unique property of Saccharomyces DHODHs could appoint DHODH as a species-specific target for novel anti-fungal therapeutics.


Subject(s)
Isoenzymes/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Saccharomyces/enzymology , Base Sequence , DNA Primers , Dihydroorotate Dehydrogenase , Electrophoresis, Polyacrylamide Gel , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Kinetics , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
12.
Protein Sci ; 13(4): 1031-42, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15044733

ABSTRACT

The flavin enzyme dihydroorotate dehydrogenase (DHOD; EC 1.3.99.11) catalyzes the oxidation of dihydroorotate to orotate, the fourth step in the de novo pyrimidine biosynthesis of UMP. The enzyme is a promising target for drug design in different biological and clinical applications for cancer and arthritis. The first crystal structure of the class 2 dihydroorotate dehydrogenase from rat has been determined in complex with its two inhibitors brequinar and atovaquone. These inhibitors have shown promising results as anti-proliferative, immunosuppressive, and antiparasitic agents. A unique feature of the class 2 DHODs is their N-terminal extension, which folds into a separate domain comprising two alpha-helices. This domain serves as the binding site for the two inhibitors and the respiratory quinones acting as the second substrate for the class 2 DHODs. The orientation of the first N-terminal helix is very different in the two complexes of rat DHOD (DHODR). Binding of atovaquone causes a 12 A movement of the first residue in the first alpha-helix. Based on the information from the two structures of DHODR, a model for binding of the quinone and the residues important for the interactions could be defined. His 56 and Arg 136, which are fully conserved in all class 2 DHODs, seem to play a key role in the interaction with the electron acceptor. The differences between the membrane-bound rat DHOD and membrane-associated class 2 DHODs exemplified by the Escherichia coli DHOD has been investigated by GRID computations of the hydrophobic probes predicted to interact with the membrane.


Subject(s)
Aniline Compounds/metabolism , Biphenyl Compounds/metabolism , Enzyme Inhibitors/metabolism , Hydroxybutyrates/metabolism , Immunosuppressive Agents/metabolism , Naphthoquinones/metabolism , Orotic Acid/analogs & derivatives , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Amino Acid Sequence , Aniline Compounds/chemistry , Animals , Atovaquone , Biphenyl Compounds/chemistry , Catalysis , Crotonates , Crystallography, X-Ray , Dihydroorotate Dehydrogenase , Drug Design , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Hydroxybutyrates/chemistry , Immunosuppressive Agents/chemistry , Models, Molecular , Molecular Sequence Data , Molecular Structure , Naphthoquinones/chemistry , Nitriles , Orotic Acid/chemistry , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Sequence Alignment , Substrate Specificity , Toluidines
13.
Arch Pharm Res ; 26(3): 197-201, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12723931

ABSTRACT

Novel beta-hydroxy propenamides as analogues of the active metabolite of leflunomide (A 771726) were synthesized and evaluated for their inhibitory activity on dihydroorotate dehydrogenase (DHODH) in an investigation into their immunosuppressive activity. Compounds 2a, 3a, and 3h were approximately 4-40 times more potent than leflunomide in their activity while they were-less active than A 771726.


Subject(s)
Aniline Compounds/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Hydroxybutyrates/chemical synthesis , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Crotonates , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydroxybutyrates/chemistry , Hydroxybutyrates/pharmacology , Isoxazoles/metabolism , Leflunomide , Nitriles , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Toluidines
14.
FEBS Lett ; 529(2-3): 346-50, 2002 Oct 09.
Article in English | MEDLINE | ID: mdl-12372626

ABSTRACT

The mitochondrial membrane bound dihydroorotate dehydrogenase (DHODH; EC 1.3.99.11) catalyzes the fourth step of pyrimidine biosynthesis. By the present correction of a known cDNA sequence for Arabidopsis thaliana DHODH we revealed the importance of the very C-terminal part for its catalytic activity and the reason why--in contrast to mammalian and insect species--the recombinant plant flavoenzyme was unaccessible to date for in vitro characterization. Structure-activity relationship studies explained that potent inhibitors of animal DHODH do not significantly affect the plant enzyme. These difference could be exploited for a novel approach to herb or pest growth control by limitation of pyrimidine nucleotide pools.


Subject(s)
Arabidopsis/enzymology , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/metabolism , Amino Acid Sequence , Base Sequence , DNA Primers , DNA, Complementary , Dihydroorotate Dehydrogenase , Electrophoresis, Polyacrylamide Gel , Humans , Kinetics , Molecular Sequence Data , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Oxidoreductases/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Substrate Specificity
15.
Insect Biochem Mol Biol ; 32(9): 1159-69, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213251

ABSTRACT

Dihydroorotate dehydrogenase (DHODH, EC 1.3.99.11), the fourth enzyme of pyrimidine de novo synthesis, is an integral flavoprotein of the inner mitchondrial membrane and is functionally connected to the respiratory chain. Here, experiments have been directed toward determining the roles of the N-terminal sequence motifs both in enzymatic properties of insect DHODH produced in vitro and the in vivo function of the protein. Full-length and three N-terminal truncated derivatives of the Drosophila melanogaster enzyme were expressed in Escherichia coli and purified. For identification on Western blots of recombinant DHODH as well as the native enzyme from flies polyclonal anti-DHODH immunoglobulins were generated and affinity-purified. The enzymatic characteristics of the four versions of DHODH were very similar, indicating that the N-terminus of the enzyme does not influence its catalytic function or its susceptibility to prominent DHODH inhibitors: A77-1726, brequinar, dichloroallyl-lawsone and redoxal. Whereas the efficacy of A77-1726 and dichloroallyl-lawsone were similar with Drosophila and human DHODH, that of brequinar and redoxal differed significantly. The differences in responses of insect DHODH and the enzyme from other species may allow the design of new agents that will selectively control insect growth, due to pyrimidine nucleotide limitation. In vivo expression of the full-length and N-truncated DHODHs from engineered transgenes revealed that the truncated proteins could not support normal de novo pyrimidine biosynthesis during development of the fly (i.e., failure to complement dhod-null mutations), apparently due to instability of the truncated proteins. It is concluded that the proper intracellular localization, directed by the N-terminal targeting and transmembrane motifs, is required for stability and subsequent proper biological function in vivo.


Subject(s)
Drosophila melanogaster/enzymology , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/metabolism , Amino Acid Sequence , Animals , Catalysis , Dihydroorotate Dehydrogenase , Drosophila melanogaster/genetics , Escherichia coli , Gene Expression , Kinetics , Molecular Sequence Data , Oxidoreductases/genetics , Oxidoreductases/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
16.
J Physiol ; 542(Pt 3): 735-41, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12154175

ABSTRACT

Diazoxide and 5-hydroxydecanoate (5-HD; C10:0) are reputed to target specifically mitochondrial ATP-sensitive K(+) (K(ATP)) channels. Here we describe K(ATP) channel-independent targets of diazoxide and 5-HD in the heart. Using submitochondrial particles isolated from pig heart, we found that diazoxide (10-100 microM) dose-dependently decreased succinate oxidation without affecting NADH oxidation. Pinacidil, a non-selective K(ATP) channel opener, did not inhibit succinate oxidation. However, it selectively inhibited NADH oxidation. These direct inhibitory effects of diazoxide and pinacidil cannot be explained by activation of mitochondrial K(ATP) channels. Furthermore, application of either diazoxide (100 microM) or pinacidil (100 microM) did not decrease mitochondrial membrane potential, assessed using TMRE (tetramethylrhodamine ethyl ester), in isolated guinea-pig ventricular myocytes. We also tested whether 5-HD, a medium-chain fatty acid derivative which blocks diazoxide-induced cardioprotection, was 'activated' via acyl-CoA synthetase (EC 6.2.1.3), an enzyme present both on the outer mitochondrial membrane and in the matrix. Using analytical HPLC and electrospray ionisation mass spectrometry, we showed that 5-HD-CoA (5-hydroxydecanoyl-CoA) is indeed synthesized from 5-HD and CoA via acyl-CoA synthetase. Thus, 5-HD-CoA may be the active form of 5-HD, serving as substrate for (or inhibiting) acyl-CoA dehydrogenase (beta-oxidation) and/or exerting some other cellular action. In conclusion, we have identified K(ATP) channel-independent targets of 5-HD, diazoxide and pinacidil. Our findings question the assumption that sensitivity to diazoxide and 5-HD implies involvement of mitochondrial K(ATP) channels. We propose that pharmacological preconditioning may be related to partial inhibition of respiratory chain complexes.


Subject(s)
Adenosine Triphosphate/physiology , Decanoic Acids/pharmacology , Diazoxide/pharmacology , Heart/drug effects , Hydroxy Acids/pharmacology , Myocardium/metabolism , Potassium Channels/physiology , Repressor Proteins , Saccharomyces cerevisiae Proteins , Acyl Coenzyme A/metabolism , Animals , Coenzyme A Ligases/metabolism , Electron Transport/drug effects , Flavoproteins/physiology , Fluorescence , Guinea Pigs , Membrane Potentials/drug effects , Mitochondria, Heart/physiology , Pinacidil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...