Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Arch Biochem Biophys ; 645: 26-33, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29524429

RESUMO

Deuterated proteins and other bio-derived molecules are important for NMR spectroscopy, neutron reflectometry, small angle neutron scattering, and neutron protein crystallography. In the current study we optimized expression media and cell culture conditions to produce high levels of 3 different deuterated human carbonic anhydrases (hCAs). The labeled hCAs were then characterized and tested for deuterium incorporation by mass spectrometry, temperature stability, and propensity to crystallize. The results show that is possible to get very good yields (>10 mg of pure protein per liter of cell culture under deuterated conditions) and that protein solubility is unaffected at the crystallization concentrations tested. Using unlabeled carbon source and recycled heavy water, we were able to get 65-77% deuterium incorporation, sufficient for most neutron-based techniques, and in a very cost-effective way. For most deuterated proteins characterized in the literature, the solubility and thermal stability is reduced. The data reported here is consistent with these observations and it was clear that there are measurable differences between hydrogenous and deuterated versions of the same protein in Tm and how they crystallize.


Assuntos
Anidrases Carbônicas/química , Meios de Cultura/química , Deutério/química , Nêutrons , Temperatura , Cristalização , Cristalografia , Estabilidade Enzimática , Humanos , Desdobramento de Proteína
2.
Acta Crystallogr D Struct Biol ; 75(Pt 10): 895-903, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31588921

RESUMO

Human carbonic anhydrase IX (CA IX) expression is upregulated in hypoxic solid tumours, promoting cell survival and metastasis. This observation has made CA IX a target for the development of CA isoform-selective inhibitors. To enable structural studies of CA IX-inhibitor complexes using X-ray and neutron crystallography, a CA IX surface variant (CA IXSV; the catalytic domain with six surface amino-acid substitutions) has been developed that can be routinely crystallized. Here, the preparation of protiated (H/H), H/D-exchanged (H/D) and deuterated (D/D) CA IXSV for crystallographic studies and their structural comparison are described. Four CA IXSV X-ray crystal structures are compared: two H/H crystal forms, an H/D crystal form and a D/D crystal form. The overall active-site organization in each version is essentially the same, with only minor positional changes in active-site solvent, which may be owing to deuteration and/or resolution differences. Analysis of the crystal contacts and packing reveals different arrangements of CA IXSV compared with previous reports. To our knowledge, this is the first report comparing three different deuterium-labelled crystal structures of the same protein, marking an important step in validating the active-site structure of CA IXSV for neutron protein crystallography.


Assuntos
Antígenos de Neoplasias/química , Anidrase Carbônica IX/química , Domínio Catalítico , Modelos Moleculares , Clonagem Molecular , Cristalografia por Raios X/métodos , Deutério , Escherichia coli/genética , Humanos
3.
J Mol Biol ; 301(4): 827-37, 2000 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-10966789

RESUMO

In contrast to all known deoxyribonucleoside kinases, a single highly efficient deoxyribonucleoside kinase from Drosophila melanogaster (Dm-dNK) is able to phosphorylate all precursor nucleosides for DNA synthesis. Dm-dNK was mutated in vitro by high-frequency random mutagenesis, expressed in the thymidine kinase-deficient Escherichia coli strain KY895 and clones were selected for sensitivity to the nucleoside analogs 1-beta-d-arabinofuranosylcytosine (AraC, Cytarabine), 3'-azido-2', 3'-dideoxythymidine (AZT, Zidovudine, Retrovir, 2', 3'-dideoxyadenosine (ddA) and 2',3'-dideoxycytidine (ddC, Zalcitabine, Hivid. Thirteen mutants with increased sensitivity compared to the wild-type Dm-dNK were isolated from a relatively small pool of less than 10,000 clones. Eight mutant Dm-dNKs increased the sensitivity of KY895 to more than one analog, and two of these mutants even to all four nucleoside analogs. Surprisingly, the mutations did not map to the five regions which are highly conserved among deoxyribonucleoside kinases. The molecular background of improved sensitivity was characterized for the double-mutant MuD (N45D, N64D), where the LD(100) value of transformed KY895 decreased 316-fold for AZT and more than 11-fold for ddC when compared to wild-type Dm-dNK. Purified recombinant MuD displayed higher K(m) values for the native substrates than wild-type Dm-dNK and the V(max) values were substantially lower. On the other hand, the K(m) and V(max) values for AZT and the K(m) value for ddC were nearly unchanged between MuD and wild-type Dm-dNK. Additionally, a decrease in feedback inhibition of MuD by thymidine triphosphate (TTP) was found. This study demonstrates how high-frequency mutagenesis combined with a parallel selection for desired properties provides an insight into the structure-function relationships of the multisubstrate kinase from D. melanogaster. At the same time these mutant enzymes exhibit properties useful in biotechnological and medical applications.


Assuntos
Drosophila melanogaster/enzimologia , Mutação/genética , Fosfotransferases (Aceptor do Grupo Álcool)/química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Citarabina/metabolismo , Didesoxiadenosina/metabolismo , Evolução Molecular Direcionada , Drosophila melanogaster/genética , Ativação Enzimática/efeitos dos fármacos , Retroalimentação/efeitos dos fármacos , Concentração Inibidora 50 , Cinética , Fosforilação/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Reação em Cadeia da Polimerase , Especificidade por Substrato , Timidina/metabolismo , Nucleotídeos de Timina/metabolismo , Nucleotídeos de Timina/farmacologia , Zalcitabina/metabolismo , Zidovudina/metabolismo
4.
FEBS Lett ; 467(1): 27-30, 2000 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-10664450

RESUMO

Mitochondrial dihydroorotate dehydrogenase (DHOdehase; EC 1.3.99.11) is a target of anti-proliferative, immunosuppressive and anti-parasitic agents. Here, redoxal, (2,2'-[3,3'-dimethoxy[1, 1'-biphenyl]-4,4'-diyl)diimino]bis-benzoic acid, was studied with isolated mitochondria and the purified recombinant human and rat enzyme to find out the mode of kinetic interaction with this target. Its pattern of enzyme inhibition was different from that of cinchoninic, isoxazol and naphthoquinone derivatives and was of a non-competitive type for the human (K(ic)=402 nM; K(iu)=506 nM) and the rat enzyme (K(ic)=116 nM; K(iu)=208 nM). The characteristic species-related inhibition of DHOdehase found with other compounds was less expressed with redoxal. In human and rat mitochondria, redoxal did not inhibit NADH-induced respiration, its effect on succinate-induced respiration was marginal. This was in contrast to the sound effect of atovaquone and dichloroallyl-lawsone, studied here for comparison. In human mitochondria, the IC(50) value for the inhibition of succinate-induced respiration by atovaquone was 6.1 microM and 27.4 microM for the DHO-induced respiration; for dichlorallyl-lawsone, the IC(50) values were 14.1 microM and 0.23 microM.


Assuntos
Compostos de Aminobifenil/farmacologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/antagonistas & inibidores , Oxirredutases/metabolismo , Compostos de Aminobifenil/química , Animais , Atovaquona , Di-Hidro-Orotato Desidrogenase , Desenho de Fármacos , Transporte de Elétrons/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Cinética , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/enzimologia , NAD/metabolismo , Naftoquinonas/química , Naftoquinonas/farmacologia , Ácido Orótico/análogos & derivados , Ácido Orótico/metabolismo , Oxirredução/efeitos dos fármacos , Ratos , Ácido Succínico/metabolismo
5.
Biochem Pharmacol ; 56(9): 1259-64, 1998 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9802339

RESUMO

The isoxazol leflunomide (N-(4-trifluoromethylphenyl)-5-methylisoxazol-4-carboxamide) and its active metabolite A77-1726 (N-(4-trifluoromethyl)-phenyl-2-cyano-3-hydroxy-crotonic acidamide) are promising disease-modifying antirheumatic drugs now in clinical trials. The malononitrilamides MNA279 (2-cyano-3-cyclopropyl-3-oxo-(4-cyanophenyl)propionamide) MNA715(N-(4-trifluoromethyl)-phenyl-2-cyano-3-hydroxy-hept-2-en-6- in-carboxylic acidamide) and HR325 (1(3-methyl-4-trifluoro methylphenyl-carbamoyl)-2-cyclopropyl-2oxo-propionitrile) were shown to block rejection after allograft and xenograft transplantation in animals. Brequinar and other cinchoninic acid derivatives have also been evaluated as immuno-suppressive agents. A77-1726, HR325 and brequinar have been shown to have strong inhibitory effects on mitochondrial dihydroorotate dehydrogenase [EC 1.3.99.11], the fourth enzyme of pyrimidine de novo synthesis, with concomitant reduction of pyrimidine nucleotide pools. Pyrimidine nucleotides are essential for normal immune cell functions. Because most investigations had been carried out with cells, cell homogenates or mitochondrial fractions, it was the rationale of the present study to differentiate, under standardized conditions, the effect of leflunomide, A77-1726, MNA279, MNA715, HR 325 and brequinar on the recombinant rat and human enzymes, which were purified in our laboratory. Whereas leflunomide was a relatively weak inhibitor of the rat (IC50 = 6.3 microM) and human (IC50 = 98 microM) dihydroorotate dehydrogenase, the influence of A77-1726, MNA 279, MNA715 and HR325 was of comparable efficacy for either the rat (range of IC50, 19-53 nM) or the human enzyme (range of IC50, 0.5-2.3 microM). From the IC50 values, it was deduced that brequinar was a more potent inhibitor of the human dihydroorotate dehydrogenase activity (IC50 = 10 nM) than of the rat enzyme (IC50 = 367 nM). The rat enzyme was influenced by all isoxazol derivatives to a greater extent (IC50 = 19 nM A77-1726) than the human enzyme (IC50 = 1.1 microM A77-1726). These results may provide a plausible explanation for the findings of other laboratories with cultured cell lines and lymphocytes: in comparison to cells derived from human tissues, rat and other rodent cells were more susceptible to the isoxazol derivatives and less susceptible to brequinar. Our detailed kinetic investigations of the bisubstrate reaction catalyzed by rat dihydroorotate dehydrogenase revealed a noncompetitive type of inhibition by A77-1726 with respect to the substrate dihydroorotate and the cosubstrates ubiquinone or decylubiquinone. For brequinar, the inhibition was noncompetitive with respect to the substrate dihydroorotate, whereas with the quinone it was found to follow the "mixed typed" inhibition. In addition, brequinar acted as a "slow-binding" inhibitor of the human dihydroorotate dehydrogenase, a feature that might be of consequence for the reversibility of the reaction with the target.


Assuntos
Androstenóis/farmacologia , Inibidores Enzimáticos/farmacologia , Imunossupressores/farmacologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/antagonistas & inibidores , Quinolinas/farmacologia , Compostos de Anilina/farmacologia , Animais , Di-Hidro-Orotato Desidrogenase , Humanos , Isoxazóis/farmacologia , Leflunomida , Ratos , Especificidade da Espécie
6.
Chem Biol Interact ; 124(1): 61-76, 2000 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-10658902

RESUMO

Mitochondrially-bound dihydroorotate dehydrogenase (EC 1.3.99.11) catalyzes the fourth sequential step in the de novo synthesis of uridine monophosphate. The enzyme has been identified as or surmised to be the pharmacological target for isoxazol, triazine, cinchoninic acid and (naphtho)quinone derivatives, which exerted antiproliferative, immunosuppressive, and antiparasitic effects. Despite this broad spectrum of biological and clinical relevance, there have been no comparative studies on drug-dihydroorotate dehydrogenase interactions. Here, we describe a study of the inhibition of the purified recombinant human and rat dihydroorotate dehydrogenase by ten compounds. 1,4-Naphthoquinone, 5,8-hydroxy-naphthoquinone and the natural compounds juglon, plumbagin and polyporic acid (quinone derivative) were found to function as alternative electron acceptors with 10-30% of control enzyme activity. The human and rat enzyme activity was decreased by 50% by the natural compound lawsone ( > 500 and 49 microM, respectively) and by the derivatives dichloroally-lawsone (67 and 10 nM), lapachol (618 and 61 nM) and atovaquone (15 microM and 698 nM). With respect to the quinone co-substrate of the dihydroorotate dehydrogenase, atovaquone (Kic = 2.7 microM) and dichloroally-lawsone (Kic = 9.8 nM) were shown to be competitive inhibitors of human dihydroorotate dehydrogenase. Atovaquone (Kic = 60 nM) was also acompetitive inhibitor of the rat enzyme. Dichloroally]-lawsone was found to be a time-dependent inhibitor of the rat enzyme, with the lowest inhibition constant (Ki* = 0.77 nM) determined so far for mammalian dihydroorotate dehydrogenases. Another inhibitor, brequinar was previously reported to be a slow-binding inhibitor of the human dihydroorotate dehydrogenase [W. Knecht, M. Loffler, Species-related inhibition of human and rat dihyroorotate dehydrogenase by immunosuppressive isoxazol and cinchoninic acid derivatives, Biochem. Pharmacol. 56 (1998) 1259-1264]. The slow binding features of this potent inhibitor (Ki* = 1.8 nM) with the human enzyme, were verified and seen to be one of the reasons for the narrow therapeutic window (efficacy versus toxicity) reported from clinical trials on its antiproliferative and immunosuppressive action. With respect to the substrate dihydroorotate, atovaquone was an uncompetitive inhibitor of human dihydroorotate dehydrogenase (Kiu = 11.6 microM) and a non-competitive inhibitor of the rat enzyme (Kiu = 905/ Kic = 1,012 nM). 1.5 mM polyporic acid, a natural quinone from fungi, influenced the activity of the human enzyme only slightly; the activity of the rat enzyme was decreased by 30%.


Assuntos
Inibidores Enzimáticos/farmacologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/antagonistas & inibidores , Animais , Atovaquona , Benzoquinonas/metabolismo , Benzoquinonas/farmacologia , Ligação Competitiva , Compostos de Bifenilo/metabolismo , Compostos de Bifenilo/farmacologia , Di-Hidro-Orotato Desidrogenase , Inibidores Enzimáticos/metabolismo , Humanos , Concentração Inibidora 50 , Cinética , Naftoquinonas/metabolismo , Naftoquinonas/farmacologia , Oxirredutases/metabolismo , Quinonas/metabolismo , Quinonas/farmacologia , Ratos , Especificidade por Substrato
7.
Nucleosides Nucleotides Nucleic Acids ; 23(8-9): 1281-5, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15571246

RESUMO

To follow the expression of the fourth enzyme of pyrimidine de novo synthesis dihydroorotate dehydrogenase (DHODH) in cells and tissues, we studied the DHODH mRNA expression by means of RT-PCR in rat tissues. Rabbit polyclonal anti-DHODH immunoglobulins were applied for immunochemical quantification of the enzyme protein by Western blotting. In mouse B-lymphocytes, which were adapted to tolerate up to a 50-fold concentration of the DHODH inhibitor leflunomide, a 20 fold protein overexpression was measured. Southern blotting indicated DHODH gene amplification.


Assuntos
Resistência a Medicamentos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Animais , Southern Blotting , Western Blotting , Linhagem Celular , DNA/química , Di-Hidro-Orotato Desidrogenase , Humanos , Imunoglobulinas/metabolismo , Imuno-Histoquímica , Concentração Inibidora 50 , Isoxazóis/farmacologia , Leflunomida , Linfócitos/metabolismo , Masculino , Membranas/metabolismo , Camundongos , Mitocôndrias/metabolismo , Pirimidinas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo , Fatores de Tempo
8.
Br J Pharmacol ; 171(16): 3814-26, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24749982

RESUMO

BACKGROUND AND PURPOSE: Although serine proteases and agonists of protease-activated receptor 2 (PAR2) cause inflammation and pain, the spectrum of proteases that are activated by proinflammatory and algesic stimuli and their contribution to inflammatory pain are uncertain. EXPERIMENTAL APPROACH: Enzymic assays and selective inhibitors were used to characterize protease activity in mice after intraplantar injections of formalin, bradykinin, PAR2 activating peptide (AP) or vehicle. The capacity of these proteases and of recombinant mouse trypsin 4 to cleave fragments of PAR2 and to activate PAR2 in cell lines was determined. Protease inhibitors and par2 (-/-) mice were used to assess the contributions of proteases and PAR2 to pain and inflammation. KEY RESULTS: Intraplantar injection of formalin, bradykinin or PAR2-AP led to the activation of proteases that were susceptible to the serine protease inhibitor melagatran but resistant to soybean trypsin inhibitor (SBTI). Melagatran inhibited mouse trypsin 4, which degraded SBTI. Proteases generated in inflamed tissues cleaved PAR2-derived peptides. These proteases and trypsin 4 increased [Ca(2+) ]i in PAR2-transfected but not in untransfected cells, and melagatran suppressed this activity. Melagatran or PAR2 deletion suppressed oedema and mechanical hypersensitivity induced by intraplantar formalin, bradykinin and PAR2-AP, but had no effect on capsaicin-induced pain. CONCLUSIONS AND IMPLICATIONS: Diverse proinflammatory and algesic agents activate melagatran-sensitive serine proteases that cause inflammation and pain by a PAR2-mediated mechanism. By inducing self-activating proteases, PAR2 amplifies and sustains inflammation and pain. Serine protease inhibitors can attenuate the inflammatory and algesic effects of diverse stimuli, representing a useful therapeutic strategy.


Assuntos
Inflamação/metabolismo , Dor/metabolismo , Receptor PAR-2/metabolismo , Serina Proteases/metabolismo , Animais , Azetidinas/farmacologia , Benzilaminas/farmacologia , Bradicinina , Linhagem Celular , Feminino , , Formaldeído , Inflamação/induzido quimicamente , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligopeptídeos , Dor/induzido quimicamente , Receptor PAR-2/agonistas , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Inibidores de Serina Proteinase/farmacologia , Tripsina/metabolismo
12.
Gene Ther ; 14(17): 1278-86, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17581598

RESUMO

Transduced deoxyribonucleoside kinases (dNK) can be used to kill recipient cells in combination with nucleoside prodrugs. The Drosophila melanogaster multisubstrate dNK (Dm-dNK) displays a superior turnover rate and has a great plasticity regarding its substrates. We used directed evolution to create Dm-dNK mutants with increased specificity for several nucleoside analogs (NAs) used as anticancer or antiviral drugs. Four mutants were characterized for the ability to sensitize Escherichia coli toward analogs and for their substrate specificity and kinetic parameters. The mutants had a reduced ability to phosphorylate pyrimidines, while the ability to phosphorylate purine analogs was relatively similar to the wild-type enzyme. We selected two mutants, for expression in the osteosarcoma 143B, the glioblastoma U-87M-G and the breast cancer MCF7 cell lines. The sensitivities of the transduced cell lines in the presence of the NAs fludarabine (F-AraA), cladribine (CdA), vidarabine and cytarabine were compared to the parental cell lines. The sensitivity of 143B cells was increased by 470-fold in the presence of CdA and of U-87M-G cells by 435-fold in the presence of F-AraA. We also show that a choice of the selection and screening system plays a crucial role when optimizing suicide genes by directed evolution.


Assuntos
Antimetabólitos , Drosophila melanogaster/enzimologia , Terapia Genética/métodos , Mutação , Neoplasias/terapia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Cladribina/uso terapêutico , Citarabina/uso terapêutico , Evolução Molecular Direcionada/métodos , Genes Transgênicos Suicidas , Glioblastoma/terapia , Humanos , Dose Letal Mediana , Osteossarcoma/terapia , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Purinas/metabolismo , Especificidade por Substrato , Transdução Genética/métodos , Vidarabina/análogos & derivados , Vidarabina/uso terapêutico
13.
Eur J Biochem ; 236(2): 609-13, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8612635

RESUMO

Mitochondrially bound dihydroorotate dehydrogenase catalyses the fourth sequential step in the de novo synthesis of uridine monophosphate. 312-bp and 983-bp regions of the human dihydroorotate dehydrogenase sequence (1496 bp) were amplified by the polymerase chain reaction, and subcloned into the expression vector pQE 32. The identity of the PCR products was verified by dideoxynucleotide sequencing. Transformation of Escherichia coli strain M15 resulted in expression of 13-kDa and 36-kDa proteins with an affinity tag consisting of six consecutive histidine residues; these proteins could be purified by solubilisation in 8 M urea and by chromatography on a Ni2+-chelating resin. In immunoblotting analyses, the fusion proteins were recognised by polyclonal avian and mammalian anti-peptide immunoglobulins. These were generated against synthetic peptides corresponding to two amino acid sequences deduced from human and rat cDNA of dihydroorotate dehydrogenase. The peptides were synthesized as multiple copies on a branching lysyl matrix. Rabbits and laying hens were immunized with these peptides without conjugation to a carrier protein. Comparison of the anti-peptide immunoglobulins produced from egg yolk and rabbit serum demonstrated that avian anti-(dihydroorotate dehydrogenase) immunoglobulins may be considered a superior alternative to the mammalian equivalent; antibodies from both sources were applicable for all immunochemical purposes. Here, these antibodies were applied for identification of a 44-kDa protein from rat liver mitochondria, which was correlated with dihydroorotate dehydrogenase activity.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/imunologia , Peptídeos/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Galinhas , Di-Hidro-Orotato Desidrogenase , Humanos , Imunoglobulina G/imunologia , Imunoglobulinas/imunologia , Dados de Sequência Molecular , Peso Molecular , Oxirredutases/química , Coelhos , Ratos , Proteínas Recombinantes
14.
Eur J Biochem ; 268(6): 1861-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11248707

RESUMO

Mammalian dihydroorotate dehydrogenase, the fourth enzyme of pyrimidine de novo synthesis is an integral protein of the inner mitochondrial membrane that faces the intermembrane space and is functionally connected to the respiratory chain via ubiquinone. Here, we describe the first cloning and analyzing of the complete cDNA of mouse dihydroorotate dehydrogenase. Based on our recent functional expression of the full-length rat and human dihydroorotate dehydrogenase, here we expressed N-terminal-truncated C-terminal-histidine-tagged constructs of the mouse, rat and human enzymes in Escherichia coli. These proteins were devoid of the N-terminal bipartite sequence consisting of the mitochondrial targeting sequence and adjacent hydrophobic domain necessary for import and proper location and fixation of the enzyme in the inner mitochondrial membrane. By employing metal-chelate affinity chromatography under native conditions, the enzymes were purified without detergents to a specific activity of more than 100 micromol x min(-1) x mg(-1) at pH optimum of 8.0--8.1. Flavin analyses by UV-visible spectrometry of the native enzymes gave fairly stoichiometric ratios of 0.6--1.2 mol flavin per mol protein. The kinetic constants of the truncated rat enzyme (K(m) = 11 microM dihydroorotate; K(m) = 7 microM ubiquinone) and human enzyme (K(m) = 10 microM dihydroorotate; K(m) = 14 microM ubiquinone) were very close to those recently reported for the full-size enzymes. The constants for the mouse enzyme, K(m) = 26 microM dihydroorotate and K(m) = 62 microM ubiquinone, were slightly elevated in comparison to those of the other species. The three truncated enzymes were tested for their efficacy with five inhibitors of topical clinical relevance against autoimmune disorders and tumors. Whereas the presence of the N-terminus of dihydroorotate dehydrogenase was essentially irrelevant for the efficacy of the malononitrilamides A77-1726, MNA715 and MNA279 with the rat and human enzyme, the N-termini were found to be important for the efficacy of the dianisidine derivative redoxal. Moreover, the complete N-terminal part of the human enzyme seemed to be of crucial importance for the 'slow-binding' features of the cinchoninic acid derivative brequinar, which was suggested to be one of the reasons for the narrow therapeutic window reported from clinical trials on its anti-proliferative and immunosuppressive action.


Assuntos
Inibidores Enzimáticos/farmacologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , DNA Complementar , Di-Hidro-Orotato Desidrogenase , Eletroforese em Gel de Poliacrilamida , Humanos , Cinética , Camundongos , Dados de Sequência Molecular , Mutação , Oxirredutases/antagonistas & inibidores , Oxirredutases/genética , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
15.
Protein Expr Purif ; 13(3): 414-22, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9693067

RESUMO

Mitochondrially bound dihydroorotate-ubiquinone oxidoreductase (dihydroorotate dehydrogenase, EC 1.3.99.11) catalyzes the fourth sequential step in the de novo synthesis of uridine monophosphate. Based on the recent functional expression of the complete rat dihydroorotate dehydrogenase by means of the baculovirus expression vector system in Trichoplusia ni cells, a procedure is described that allows the purification of baculovirus expressed enzyme protein fused to a carboxy-terminal tag of eight histidines. Extracts from mitochondria of Spodoptera frugiperda cells infected with the recombinant virus using Triton X-100 were loaded onto Ni2+-nitrilotriacetic acid agarose and histidine-tagged rat protein was selectively eluted with imidazole-containing buffer. In view of our previously published work, the quality of the electrophoretic homogenous rat enzyme was markedly improved; specific activity was 130-150 micromol dihydroorotate/min per milligram; and the stoichiometry of flavin content was 0.8-1.1 mol/mol protein. Efforts to generate mammalian dihydroorotate dehydrogenases with low production costs from bacteria resulted in successful overexpression of the carboxy-terminal-modified rat and human dihydroorotate dehydrogenase in XL-1 Blue cells. By employing the metal chelate affinity chromatography under native conditions, the histidine-tagged human enzyme was purified with a specific activity of 150 micromol/min/mg and the rat enzyme with 83 micromol/min/mg, respectively, at pH 8.0-8.1 optimum. Kinetic constants of the recombinant histidine-tagged rat enzyme from bacteria (dihydroorotate, Km = 14.6 micromol electron acceptor decylubiquinone, Km = 9.5 micromol) were close to those reported for the enzyme from insect cells, with or without the affinity tag. HPLC analyses identified flavin mononucleotide as cofactor of the rat enzyme; UV-vis and fluorometric analyses verified a flavin/protein ratio of 0.8-1.1 mol/mol. By spectral analyses of the functional flavin with the native human enzyme, the interaction of the pharmacological inhibitors Leflunomide and Brequinar with their target could be clarified as interference with the transfer of electrons from the flavin to the quinone. The combination of the bacterial expression system and metal chelate affinity chomatography offers an improved means to purify large quantities of mammalian membrane-bound dihydroorotate dehydrogenases which, by several criteria, possesses the same functional activities as non-histidine-tagged recombinant enzymes.


Assuntos
Histidina/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/genética , Animais , Baculoviridae/genética , DNA Complementar , Di-Hidro-Orotato Desidrogenase , Eletroforese em Gel de Poliacrilamida , Humanos , Concentração de Íons de Hidrogênio , Cinética , Dados de Sequência Molecular , Oxirredutases/isolamento & purificação , Oxirredutases/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Spodoptera
16.
Biol Cybern ; 66(4): 327-33, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1550882

RESUMO

Existing computational models of structure-from-motion--the appearance of three-dimensional motion generated by moving two-dimensional patterns--are all based on variations of optical flow or feature point correspondence within the interior of single objects. Three separate phenomena provide strong evidence that in human vision, structure-from-motion is significantly affected by surface boundary cues. In the first, a rotating cylinder is seen, though no variation in optical flow exists across the apparent cylinder. In the second, the shape of the bounding contour of a moving pattern dominates the actual differential motion within the pattern. In the third, the appearance of independently moving objects changes significantly when the boundary between them becomes indistinct. We describe a simple computational model sufficient to account for these effects. The model is based on qualitative constraints relating possible object motions to patterns of flow, together with an understanding of the patterns of flow that can be discriminated in practice.


Assuntos
Percepção Visual/fisiologia , Animais , Humanos , Matemática , Modelos Biológicos , Rotação
17.
Eur J Biochem ; 267(7): 2079-87, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10727948

RESUMO

In animals, dihydroorotate dehydrogenase (DHODH) is a mitochondrial protein that carries out the fourth step in de novo pyrimidine biosynthesis. Because this is the only enzyme of this pathway that is localized to mitochondria and because the enzyme is cytosolic in some bacteria and fungi, we carried out studies to understand the mode of targeting of animal DHODH and its submitochondrial localization. Analysis of fractionated rat liver mitochondria revealed that DHODH is an integral membrane protein exposed to the intermembrane space. In vitro-synthesized Drosophila, rat and human DHODH proteins were efficiently imported into the intermembrane space of isolated yeast mitochondria. Import did not alter the size of the in vitro synthesized protein, nor was there a detectable size difference when compared to the DHODH protein found in vivo. Thus, there is no apparent proteolytic processing of the protein during import either in vitro or in vivo. Import of rat DHODH into isolated yeast mitochondria required inner membrane potential and was at least partially dependent upon matrix ATP, indicating that its localization uses the well described import machinery of the mitochondrial inner membrane. The DHODH proteins of animals differ from the cytosolic proteins found in some bacteria and fungi by the presence of an N-terminal segment that resembles mitochondrial-targeting presequences. Deletion of the cationic portion of this N-terminal sequence from the rat DHODH protein blocked its import into isolated yeast mitochondria, whereas deletion of the adjacent hydrophobic segment resulted in import of the protein into the matrix. Thus, the N-terminus of the DHODH protein contains a bipartite signal that governs import and correct insertion into the mitochondrial inner membrane.


Assuntos
Proteínas de Membrana/metabolismo , Mitocôndrias/enzimologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Primers do DNA , Di-Hidro-Orotato Desidrogenase , Humanos , Hidrólise , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Oxirredutases/química , Oxirredutases/genética , Saccharomyces cerevisiae/enzimologia
18.
Protein Expr Purif ; 10(1): 89-99, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9179295

RESUMO

Mammalian dihydroorotate dehydrogenase (EC 1.3.99.11), the fourth enzyme of pyrimidine de novo synthesis is located in the mitochondrial inner membrane with functional connection to the respiratory chain. From the cDNA of rat liver dihydroorotate dehydrogenase cloned in our laboratory the first complete sequence of a mammalian enzyme was deduced. Two hydrophobic stretches centered around residues 20 and 357, respectively, and a short N-terminal mitochondrial targeting sequence of 10 amino acids was proposed. A recombinant baculovirus containing the rat liver cDNA for dihydroorotate dehydrogenase was constructed and used for virus infection and protein expression in Trichoplusia ni cells. The targeting of the recombinant protein to mitochondria of the insect cells was monitored by activity determination of dihydroorotate dehydrogenase in subcellular compartments in comparison to succinate dehydrogenase activity (EC 1.3.5.1), which is a specific marker enzyme of the inner mitochondrial membrane. The results of subcellular distribution were verified by Western blotting with anti-dihydroorotate dehydrogenase immunoglobulins. The activity of the recombinant enzyme in the mitochondria of infected insect cells was found to be about 570-fold above the level of dihydroorotate dehydrogenase in rat liver mitochondria. By cation exchange chromatography of the Triton X-114 solubilisate of mitochondria, dihydroorotate dehydrogenase was purified to give a specific activity of 15 U/mg at pH 8.0. This was a marked progress over the six-step purification procedure of the enzyme from rat liver which resulted in a specific activity of 0.7 U/mg at pH 8.0. The characteristic flavin absorption spectrum obtained with the recombinant enzyme gave strong evidence that the rodent enzyme is a flavoprotein. By enzyme kinetic studies K(m) values for dihydroorotate and ubiquinone were 6.4 and 9.9 microM with the recombinant enzyme, and were 5.0 and 19.7 microM, respectively, with the rat liver enzyme. After expression of only truncated forms of human dihydroorotate dehydrogenase, the present successful generation of the complete rodent enzyme using insect cells and the efficient procedure will promote structure and function studies of the eukaryotic dihydroorotate dehydrogenases in comparison to the microbial enzyme.


Assuntos
Mitocôndrias/química , Mariposas/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/genética , Ratos/genética , Sequência de Aminoácidos , Animais , Western Blotting , Linhagem Celular , Cromatografia por Troca Iônica , DNA Complementar/genética , Di-Hidro-Orotato Desidrogenase , Vetores Genéticos/genética , Humanos , Cinética , Fígado/química , Dados de Sequência Molecular , Mariposas/citologia , Nucleopoliedrovírus/genética , Ácido Orótico/análogos & derivados , Ácido Orótico/metabolismo , Oxirredutases/biossíntese , Oxirredutases/isolamento & purificação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Spodoptera/citologia , Spodoptera/metabolismo , Especificidade por Substrato , Ubiquinona/metabolismo
19.
Eur J Biochem ; 240(1): 292-301, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8925840

RESUMO

Human mitochondrial dihydroorotate dehydrogenase (the fourth enzyme of pyrimidine de novo synthesis) has been overproduced by means of a recombinant baculovirus that contained the human cDNA fragment for this protein. After virus infection and protein expression in Trichoplusia ni cells (BTI-Tn-5B1-4), the subcellular distribution of the recombinant dihydroorotate dehydrogenase was determined by two distinct enzyme-activity assays and by Western blot analysis with anti-(dihydroorotate dehydrogenase) Ig. The targeting of the recombinant protein to the mitochondria of the insect cells was verified. The activity of the recombinant enzyme in the mitochondria of infected cells was about 740-fold above the level of dihydroorotate dehydrogenase in human liver mitochondria. In a three-step procedure, dihydroorotate dehydrogenase was purified to a specific activity of greater than 50 U/mg. Size-exclusion chromatography showed a molecular mass of 42 kDa and confirmed the existence of the fully active enzyme as a monomeric species. Fluorimetric cofactor analysis revealed the presence of FMN in recombinant dihydroorotate dehydrogenase. By kinetics analysis, Km values for dihydroorotate and ubiquinone-50 were found to be 4 microM and 9.9 microM, respectively, while Km values for dihydroorotate and decylubiquinone were 9.4 microM and 13.7 microM, respectively. The applied expression system will allow preparation of large quantities of the enzyme for structure and function studies. Purified recombinant human dihytdroorotate dehydrogenase was tested for its sensitivity to a reported inhibitor A77 1726 (2-hydroxyethyliden-cyanoacetic acid 4-trifluoromethyl anilide), which is the active metabolite of the isoxazole derivative leflunomide [5-methyl-N-(4-trifluoromethyl-phenyl)-4-isoxazole carboximide]. An IC50 value of 1 microM was determined for A77 1726. Detailed kinetics experiments revealed uncompetitive inhibition with respect to dihydroorotate (Kiu = 0.94 microM) and non-competitive inhibition with respect to decylubiquinone (Kic = 1.09 microM, Kiu = 1.05 microM). These results suggest that the immunomodulating agent A77 1726 (currently in clinical phase III studies for the treatment of rheumatoid arthritis) is a very good inhibitor of human dihydroorotate dehydrogenase.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/isolamento & purificação , Oxirredutases/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Cromatografia em Gel , Di-Hidro-Orotato Desidrogenase , Eletroforese em Gel de Poliacrilamida , Flavinas/análise , Humanos , Insetos , Cinética , Mitocôndrias/enzimologia , Dados de Sequência Molecular , Peso Molecular , Nucleopoliedrovírus , Organelas/enzimologia , Oxirredutases/biossíntese , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/isolamento & purificação , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Espectrofotometria , Frações Subcelulares/enzimologia , Especificidade por Substrato , Transfecção
20.
J Biol Chem ; 275(9): 6673-9, 2000 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-10692477

RESUMO

The occurrence of a deoxyribonucleoside kinase in Drosophila melanogaster (Dm-dNK) with remarkably broad substrate specificity has recently been indicated (Munch-Petersen, B., Piskur, J., and Søndergaard, L. (1998) J. Biol. Chem. 273, 3926-3931). To prove that the capacity to phosphorylate all four deoxyribonucleosides is in fact associated to one polypeptide chain, partially sequenced cDNA clones, originating from the Berkeley Drosophila genome sequencing project, were searched for homology with human deoxyribonucleoside kinases. The total sequence of one cDNA clone and the corresponding genomic DNA was determined and expressed in Escherichia coli as a glutathione S-transferase fusion protein. The purified and thrombin cleaved recombinant protein phosphorylated the four deoxyribonucleosides with high turnover and K(m) values similar to those of the native Dm-dNK, as well as the four ribonucleosides and many therapeutical nucleoside analogs. Dm-dNK has apparently the same origin as the mammalian kinases, thymidine kinase 2, deoxycytidine kinase, deoxyguanosine kinase, and the herpes viral thymidine kinases, but it has a unique C terminus that seems to be important for catalytic activity and specificity. The C-terminal 20 amino acids were dispensable for phosphorylation of deoxyribonucleosides but necessary for full activity with purine ribonucleosides. Removal of the C-terminal 20 amino acids increased the specific activity 2-fold, but 99% of the activity was lost after removal of the C-terminal 30 amino acids.


Assuntos
Drosophila melanogaster/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Aminoácidos/análise , Animais , Clonagem Molecular , Desoxirribonucleosídeos/metabolismo , Escherichia coli , Cinética , Dados de Sequência Molecular , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/química , Proteínas Recombinantes de Fusão , Alinhamento de Sequência , Deleção de Sequência , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa