Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Syst Evol Microbiol ; 62(Pt 1): 188-195, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21378133

RESUMO

In many prokaryotic species, 16S rRNA genes are present in multiple copies, and their sequences in general do not differ significantly owing to concerted evolution. At the time of writing, the genus Haloarcula of the family Halobacteriaceae comprises nine species with validly published names, all of which possess two to four highly heterogeneous 16S rRNA genes. Existence of multiple heterogeneous 16S rRNA genes makes it difficult to reconstruct a biological phylogenetic tree using their sequence data. If the orthologous gene is able to be discriminated from paralogous genes, a tree reconstructed from orthologous genes will reflect a simple biological phylogenetic relationship. At present, however, we have no means to distinguish the orthologous rRNA operon from paralogous ones in the members of the family Halobacteriaceae. In this study, we found that the dihydroorotate oxidase gene, pyrD, was present in the immediate upstream of one 16S rRNA gene in each of ten strains of the family Halobacteriaceae whose genome sequences have been determined, and the direction of the pyrD gene was opposite to that of the 16S rRNA genes. In two other strains whose genome sequences have been determined, the pyrD gene was present in far separated positions. We designed PCR primer sets to amplify DNA fragments encompassing a region from the conserved region of the pyrD gene to a conserved region of the tRNA-Ala gene or the 23S rRNA gene to determine the 16S rRNA gene sequences preceded by the pyrD gene, and to see if the pyrD gene is conserved in the immediate upstream of rRNA operon(s) in the type strains of the type species of 28 genera of the family Halobacteriaceae. Seventeen type strains, including the ten strains mentioned above, gave amplified DNA fragments of approximately 4000 bp, while eleven type strains, including the two strains mentioned above, did not give any PCR products. These eleven strains are members of the Clade I haloarchaea, originally defined by Walsh et al. (2004) and expanded by Minegishi et al. (2010). Analysis of contig sequences of three strains belonging to the Clade I haloarchaea also revealed the absence of the pyrD gene in the immediate upstream of any 16S rRNA genes. It may be scientifically sound to hypothesize that during the evolution of members of the family Halobacteriaceae, a pyrD gene transposition event happened in one group and this was followed by subsequent speciation processes in each group, yielding species/genera of the Clade I group and 'the rest' of the present family Halobacteriaceae.


Assuntos
Di-Hidrorotato Oxidase/genética , Halobacteriaceae/classificação , Halobacteriaceae/genética , RNA Ribossômico 16S/genética , Proteínas de Bactérias/genética , Análise por Conglomerados , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Ribossômico/química , DNA Ribossômico/genética , Dados de Sequência Molecular , Filogenia , Análise de Sequência de DNA
2.
Transpl Immunol ; 23(4): 180-4, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20553870

RESUMO

We investigated the immunosuppressive effects of the dihydroortate dehydrogenase (DHODH) inhibitor compounds ABR-222417 and ABR-224050 from Active Biotech (Sweden). We verified the inhibitory effects of these compounds on the proliferation of CD4(+) and CD8(+) T-cells in vivo by using superantigen staphylococcus enterotoxin A (SEA)-mediated proliferation test. To evaluate their efficacy, the compounds were screened in a low-responder heart allograft transplantation model in rats [heart from Piebald Virol Glaxo (PVG) transplanted to Dark Agouti (DA)]. The immunosuppressive effects of the compounds were then investigated in a high-responder model (DA to PVG). Treatment with ABR-222417 (30 mg/kg) was more efficient than that with ABR-224050 (10 mg/kg), and the former provided a longer graft median survival time (MST, 29.5 days) than the latter (MST, 18.5 days). Furthermore, there was a marked increase in graft survival time (53 days) when low doses of ABR-222417 and cyclosporine A (CsA) were used in combination. No sign of tolerability problems was detected using this combination or when ABR-222417 was used singly at a higher dose. Furthermore, T-cell proliferation studies in vitro support that the anti proliferative effect of ABR-222417 is caused by inhibition of de novo pyrimidine synthesis, which is the consequence of DHODH inhibition. These results show that ABR-222417 had marked immunosuppressive effects on the heart allograft transplantation and that it exerts an even more powerful inhibitory effect on graft rejection when used in combination with CsA, with good tolerability.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Coração , ortoaminobenzoatos/administração & dosagem , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Proliferação de Células/efeitos dos fármacos , Ciclosporina/administração & dosagem , Di-Hidrorotato Oxidase/antagonistas & inibidores , Sinergismo Farmacológico , Quimioterapia Combinada , Sobrevivência de Enxerto/imunologia , Humanos , Terapia de Imunossupressão , Células Jurkat , Masculino , Ratos , Ratos Endogâmicos , ortoaminobenzoatos/farmacologia
3.
Bioorg Med Chem Lett ; 15(21): 4854-7, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16143532

RESUMO

Novel DHODH inhibitors were developed based on a previously described series by introduction of heteroatoms into the cyclopentene ring and hydroxyl groups attached to it. Also, the hydrophobic biphenyl side chain was replaced with benzyloxy phenyl groups. Activities on human, rat, and mouse enzymes indicate a species specificity of these inhibitors.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ciclopentanos/síntese química , Di-Hidrorotato Oxidase/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Imunossupressores/síntese química , Amidas , Animais , Ciclopentanos/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Imunossupressores/farmacologia , Concentração Inibidora 50 , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Camundongos , Ratos , Especificidade da Espécie , Relação Estrutura-Atividade
4.
Plant Physiol ; 138(4): 1926-38, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16024685

RESUMO

Pyrimidines are particularly important in dividing tissues as building blocks for nucleic acids, but they are equally important for many biochemical processes, including sucrose and cell wall polysaccharide metabolism. In recent years, the molecular organization of nucleotide biosynthesis in plants has been analyzed. Here, we present a functional analysis of the pyrimidine de novo synthesis pathway. Each step in the pathway was investigated using transgenic plants with reduced expression of the corresponding gene to identify controlling steps and gain insights into the phenotypic and metabolic consequences. Inhibition of expression of 80% based on steady-state mRNA level did not lead to visible phenotypes. Stepwise reduction of protein abundance of Asp transcarbamoylase or dihydro orotase resulted in a corresponding inhibition of growth. This was not accompanied by pleiotropic effects or by changes in the developmental program. A more detailed metabolite analysis revealed slightly different responses in roots and shoots of plants with decreased abundance of proteins involved in pyrimidine de novo synthesis. Whereas in leaves the nucleotide and amino acid levels were changed only in the very strong inhibited plants, the roots show a transient increase of these metabolites in intermediate plants followed by a decrease in the strong inhibited plants. Growth analysis revealed that elongation rates and number of organs per plant were reduced, without large changes in the average cell size. It is concluded that reduced pyrimidine de novo synthesis is compensated for by reduction in growth rates, and the remaining nucleotide pools are sufficient for running basic metabolic processes.


Assuntos
/enzimologia , Pirimidinas/biossíntese , Solanum tuberosum/enzimologia , Aspartato Carbamoiltransferase/genética , Aspartato Carbamoiltransferase/metabolismo , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/genética , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/metabolismo , Di-Hidro-Orotase/genética , Di-Hidro-Orotase/metabolismo , Di-Hidrorotato Oxidase/genética , Di-Hidrorotato Oxidase/metabolismo , Regulação da Expressão Gênica de Plantas , Modelos Químicos , Dados de Sequência Molecular , Estrutura Molecular , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Orotato Fosforribosiltransferase/genética , Orotato Fosforribosiltransferase/metabolismo , Orotidina-5'-Fosfato Descarboxilase/genética , Orotidina-5'-Fosfato Descarboxilase/metabolismo , Folhas de Planta/genética , Folhas de Planta/fisiologia , Raízes de Plantas/genética , Raízes de Plantas/fisiologia , Brotos de Planta/genética , Brotos de Planta/fisiologia , Plantas Geneticamente Modificadas , Pirimidinas/química , Especificidade da Espécie
5.
Mol Biochem Parasitol ; 122(2): 189-200, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12106873

RESUMO

Trypanosoma cruzi, a protozoan causing Chagas' disease, excretes a considerable amount of succinate even though it uses the TCA cycle and the aerobic respiratory chain. For this reason, it was believed that unknown metabolic pathways participate in succinate production in this parasite. In the present study, we examined the molecular properties of dihydroorotate dehydrogenase (DHOD), the fourth enzyme of de novo pyrimidine biosynthetic pathway, as a soluble fumarate reductase (FRD) because our sequence analysis of pyr genes cluster showed that the amino acid sequence of T. cruzi DHOD is quite similar to that of type 1A DHOD of Saccharomyces cerevisiae, an enzyme that uses fumarate as an electron acceptor and produces succinate. Biochemical analyses of the cytosolic enzyme purified from the parasite and of the recombinant enzyme revealed that T. cruzi DHOD has methylviologen-fumarate reductase (MV-FRD) activity. In addition, T. cruzi DHOD was found to catalyze electron transfer from dihydroorotate to fumarate by a ping-pong Bi-Bi mechanism. The recombinant enzyme contained FMN as a prosthetic group. Dynamic light scattering analysis indicated that T. cruzi DHOD is a homodimer. These results clearly indicated that the cytosolic MV-FRD is attributable to T. cruzi DHOD. The DHOD may play an important role in succinate/fumarate metabolism as well as de novo pyrimidine biosynthesis in T. cruzi.


Assuntos
Di-Hidrorotato Oxidase/metabolismo , Paraquat/metabolismo , Succinato Desidrogenase/metabolismo , Trypanosoma cruzi/enzimologia , Animais , Sítios de Ligação , Citosol/enzimologia , Di-Hidrorotato Oxidase/genética , Di-Hidrorotato Oxidase/isolamento & purificação , Fumaratos , Cinética , Oxirredução , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidade , Succinato Desidrogenase/genética , Succinato Desidrogenase/isolamento & purificação , Succinatos/metabolismo , Trypanosoma cruzi/genética , Trypanosoma cruzi/crescimento & desenvolvimento
6.
J Med Chem ; 44(18): 2861-4, 2001 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-11520193

RESUMO

Dihydroorotate dehydrogenases (DHODs) catalyze the conversion of dihydroorotate to orotate in de novo pyrimidine biosynthesis. We have found that 3,4-dihydroxybenzoate and 3,5-dihydroxybenzoate are competitive inhibitors vs dihydroorotate with the prototypical family 1A DHOD from Lactococcus lactis. The dissociation constants of these compounds, determined by spectral titrations, were similar to the dissociation constant of orotate, the enzymatic reaction product, suggesting that hydroxybenzoates could be developed into useful drugs for treating infections by certain protozoan parasites.


Assuntos
Benzoatos/síntese química , Di-Hidrorotato Oxidase/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Pirimidinas/síntese química , Tripanossomicidas/síntese química , Benzoatos/química , Inibidores Enzimáticos/química , Ligantes , Pirimidinas/química , Relação Estrutura-Atividade , Tripanossomicidas/química
7.
Ann Rheum Dis ; 59(11): 841-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11053058

RESUMO

Leflunomide is a selective inhibitor of de novo pyrimidine synthesis. In phase II and III clinical trials of active rheumatoid arthritis, leflunomide was shown to improve primary and secondary outcome measures with a satisfactory safety profile. The active metabolite of leflunomide, A77 1726, at low, therapeutically applicable doses, reversibly inhibits dihydroorotate dehydrogenase (DHODH), the rate limiting step in the de novo synthesis of pyrimidines. Unlike other cells, activated lymphocytes expand their pyrimidine pool by approximately eightfold during proliferation; purine pools are increased only twofold. To meet this demand, lymphocytes must use both salvage and de novo synthesis pathways. Thus the inhibition of DHODH by A77 1726 prevents lymphocytes from accumulating sufficient pyrimidines to support DNA synthesis. At higher doses, A77 1726 inhibits tyrosine kinases responsible for early T cell and B cell signalling in the G(0)/G(1) phase of the cell cycle. Because the immunoregulatory effects of A77 1726 occur at doses that inhibit DHODH but not tyrosine kinases, the interruption of de novo pyrimidine synthesis may be the primary mode of action. Recent evidence suggests that the observed anti-inflammatory effects of A77 1726 may relate to its ability to suppress interleukin 1 and tumour necrosis factor alpha selectively over their inhibitors in T lymphocyte/monocyte contact activation. A77 1726 has also been shown to suppress the activation of nuclear factor kappaB, a potent mediator of inflammation when stimulated by inflammatory agents. Continuing research indicates that A77 1726 may downregulate the glycosylation of adhesion molecules, effectively reducing cell-cell contact activation during inflammation.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Artrite Reumatoide/tratamento farmacológico , Isoxazóis/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Divisão Celular/efeitos dos fármacos , DNA/biossíntese , DNA/efeitos dos fármacos , Di-Hidrorotato Oxidase/efeitos dos fármacos , Di-Hidrorotato Oxidase/metabolismo , Humanos , Interleucina-1/fisiologia , Isoxazóis/uso terapêutico , Leflunomida , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/biossíntese , Fator de Necrose Tumoral alfa/efeitos dos fármacos
8.
J Mol Evol ; 44(4): 361-70, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9089075

RESUMO

Amino acid substitution tables are essential for the proper alignment of protein sequences, and alignment scores based on them can be transformed into distance measures by various means. In the simplest case, the negative log of the score is used. This Poisson relationship assumes that all sites are equally likely to change, however. A more accurate relationship would correct for different rates of change at each residue position. Recently, Grishin (J. Mol. Evol. 41:675-679, 1995) published a set of simple equations that correct for various circumstances, including different rates of change at different sites. We have used these equations in conjunction with similarity scores that take into account constraints on amino acid interchange. Simulation studies show a linear relationship between these calculated distances and the numbers of allowed mutations based on the observed variation of rate at all sites in various proteins.


Assuntos
Sequência de Aminoácidos , Evolução Biológica , Enzimas/química , Tempo , Animais , Simulação por Computador , Di-Hidrorotato Oxidase/química , Enzimas/genética , Gliceraldeído-3-Fosfato Desidrogenases/química , Mutação , Fosfoglicerato Quinase/química , Filogenia , Distribuição de Poisson , Probabilidade , Alinhamento de Sequência , Especificidade da Espécie , Superóxido Dismutase/química , Triose-Fosfato Isomerase/química
9.
Histochem Cell Biol ; 105(2): 119-28, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8852433

RESUMO

Dihydroorotate dehydrogenase (EC 1.3.3.1 or EC 1.3.99.11) catalyzes the fourth sequential step in the de novo synthesis of uridine monophosphate. In eukaryotes it is located in the inner mitochondrial membrane, with ubiquinone as the proximal and cytochrome oxidase as the ultimate electron transfer system, whereas the rest of pyrimidine biosynthesis takes place in the cytosol. Here, the distribution of dihydroorotate dehydrogenase activity in cryostat sections of various rat tissues, and tissue samples of human skin and kidney, was visualized by light microscopy using the nitroblue tetrazolium technique. In addition, a hydrogen peroxide-producing oxidase side-reactivity of dihydroorotate dehydrogenase could be visualized by trapping the peroxide with cerium-diaminobenzidine. The pattern of activity was similar to that of succinate dehydrogenase, but revealed a less intensive staining. High activities of dihydroorotate dehydrogenase were found in tissues with known proliferative, regenerative, absorptive or excretory activities, e.g., mucosal cells of the ileum and colon crypts in the gastrointestinal tract, cultured Ehrlich ascites tumor cells, and proximal tubules of the kidney cortex, whilst lower activities were present in the periportal area of the liver, testis and spermatozoa, prostate and other glands, and skeletal muscle. Dihydroorotate dehydrogenase and succinate dehydrogenase activity in Ehrlich ascites tumor cells grown in suspension culture were quantified by application of nitroblue tetrazolium or cyanotolyl tetrazolium and subsequent extraction of the insoluble formazans with organic solvents. The ratio of dihydroorotate dehydrogenase to succinate dehydrogenase activity was 1:4. This was in accordance with that of 1:5 obtained from oxygen consumption measurement of isolated mitochondria on addition of dihydroorotate or succinate. The ratio determined with mitochondria from animal tissues was up to 1:15 (rat liver, bovine heart). The application of the enzyme inhibitors brequinar sodium and toltrazuril verified the specificity of the histochemical and biochemical methods applied.


Assuntos
Células/enzimologia , Di-Hidrorotato Oxidase/análise , Histocitoquímica/métodos , Mitocôndrias/enzimologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Oxirredutases/análise , Succinato Desidrogenase/análise , Animais , Carcinoma de Ehrlich/metabolismo , Bovinos , Di-Hidro-Orotato Desidrogenase , Feminino , Cobaias , Humanos , Masculino , Camundongos , Pirimidinas/química , Ratos , Ratos Wistar , Succinato Desidrogenase/metabolismo , Suínos
10.
Biochem Pharmacol ; 50(8): 1157-64, 1995 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-7488229

RESUMO

Penclomedine (PEN) is a synthetic pyridine derivative that has been selected for clinical development based on its activity against human and mouse breast tumors implanted in mice. Its mechanism of action was unclear, and we were interested in determining its mechanism of cytotoxicity in vitro and in vivo. We found chromosome breaks, gaps, and exchanges in P388 ascites cells from BD2F1 mice treated with 200 mg/kg PEN. Maximal observed damage occurred 24 hr after drug administration. Alkaline elution indicated only limited DNA strand breaks and interstrand cross-linking. In vitro, PEN (75 micrograms/mL) inhibited RNA and DNA syntheses almost completely. In addition, incubation of [14C]PEN with rat liver S-9 fraction in the presence of calf thymus DNA resulted in the stable transfer of radioactivity to DNA. Addition of butylated hydroxytoluene, a free radical scavenger, to the incubation mixture inhibited the binding of drug to DNA, implicating free radicals as the ultimate reactive species. These data suggest that PEN can be metabolized to free radical, DNA-reactive products, and that its cytotoxicity is due to chromosomal damage produced by monofunctional alkylation. As an alternate mechanism, the ability of PEN to inhibit cellular dihydroorotate dehydrogenase was explored. Although PEN is an inhibitor of this enzyme in cells in vivo, in vitro, and in isolated cell sonicates, HPLC analyses of ribonucleotide triphosphate pools in P388 cells showed that all triphosphates had increased, especially UTP. Addition of uridine to the cell culture failed to prevent PEN-mediated cytotoxicity, suggesting that inhibition of de novo pyrimidine biosynthesis was not likely to be an important mechanism of action of this drug. These data suggest that PEN is activated in cells to a free radical that binds DNA.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA , Picolinas/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , DNA/metabolismo , Di-Hidrorotato Oxidase/antagonistas & inibidores , Humanos , Leucemia P388 , Malondialdeído/análise , Picolinas/metabolismo , Uridina/metabolismo
11.
Histochem Cell Biol ; 103(4): 287-92, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7648404

RESUMO

Biochemical studies have demonstrated that dihydroorotate dehydrogenase (DHOdehase; EC 1.3.3.1 or 1.3.99.11) is the sole enzyme of de novo pyrimidine synthesis in mitochondria, whereas the rest of the pathway takes place in the cytosol. The dehydrogenation of dihydroorotate to orotate is linked to the respiratory chain via ubiquinone. In this study, we show for the first time the ultrastructural localization of DHOdehase. Since the purified enzyme was found to act both as dehydrogenase and as oxidase, the cerium capture technique for detecting enzymatically generated hydrogen peroxide could be applied to pin-point the in situ activity of DHOdehase oxidase in mitochondria of rat heart and kidney cortex. Cerium perhydroxide as the final reaction product was detected predominantly in the matrix with some focal condensation along the inner membrane, but not in the intermembrane space. From this pattern of localization, it is concluded that the active site of the membrane-bound enzyme could face the mitochondrial matrix similar to succinate dehydrogenase. The reliability of the applied method for the demonstration of DHOdehase oxidase was demonstrated by the addition of Brequinar sodium to the incubation medium. This quinoline-carboxylic acid derivative is a potent inhibitor of DHOdehase and has proven anti-proliferative activity. The present observations do not ascertain whether the oxidase is permanently active as a constant portion of the enzyme in vivo, similar to xanthine oxidase/dehydrogenase. However, DHOdehase should be considered as a source of radical oxygen species under pathophysiological conditions.


Assuntos
Di-Hidrorotato Oxidase/análise , Córtex Renal/enzimologia , Miocárdio/enzimologia , Animais , Compartimento Celular , Córtex Renal/ultraestrutura , Masculino , Microscopia Eletrônica , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Cardíacas/ultraestrutura , Ratos , Ratos Sprague-Dawley
13.
Antiviral Res ; 20(1): 71-81, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8384434

RESUMO

CL 306,293, a substituted quinoline carboxylic acid, is a potent inhibitor of dihydroorotic acid dehydrogenase, an enzyme essential for the biosynthesis of pyrimidines. In mammalian cell culture, the agent exhibits antiproliferative properties that can be reversed by the addition of uridine. CL 306,293 inhibits the development of the clinical disease in a murine model of immunodeficiency induced by a mixture of LP-BM5 retroviruses. In infected mice, the agent prevents the development of hypergammaglobulinemia, lymphadenopathy, splenomegaly and induction of an IL-2 deficiency. The CD4/CD8 ratio and the number of B cells in the lymph nodes are decreased if the infected animals are treated with CL 306,293. CL 306,293 was more efficacious and potent than 3'-azido-3'-deoxythymidine. The beneficial effects of CL 306,293 observed in this model are most probably related to its antiproliferative properties.


Assuntos
Aminoquinolinas/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Di-Hidrorotato Oxidase/antagonistas & inibidores , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Complexo Relacionado com a AIDS , Aminoquinolinas/toxicidade , Animais , Anticorpos Antivirais/sangue , Linfócitos B/fisiologia , Compostos de Bifenilo/toxicidade , Relação CD4-CD8 , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Hipergamaglobulinemia , Imunoglobulina M/análise , Interleucina-2/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/enzimologia , Recidiva , Esplenomegalia , Zidovudina/uso terapêutico
14.
Biochem Pharmacol ; 43(5): 1025-30, 1992 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-1313236

RESUMO

To verify the assumption of a specific and potent drug action on de novo pyrimidine biosynthesis, isolated dihydroorotate dehydrogenase (DHO-DH) (EC 1.3.3.1) was exposed to Brequinar Sodium (6-fluoro-2-(2'-fluoro-1,1'-biphenyl-4-yl)-3-methyl-4-quinoline carboxylic acid sodium salt, NSC 368 390) (Brequinar). The membrane-bound DHO-DH was purified to apparent homogeneity (25,000-fold) from rat liver mitochondria in six steps via detergent extraction and subsequent chromatography using the dye ligand Matrex Gel Orange A. Using molecular mechanistic studies (MM2) this ligand was found to mimic closely the stereochemical conformation of Brequinar. SDS-PAGE revealed two protein bands for the purified enzyme with apparent molecular masses of 58 (major) and 68 kDa (minor). In vitro, two modes of action of the DHO-DH are possible: (i) acting as a dehydrogenase in the presence of ubiquinone as proximal electron acceptor and (ii) direct reaction with oxygen as oxidase. A novel assay for the measurement of the oxidase activity was adapted using leuco-dichlorofluorescein-diacetate. Inhibition experiments revealed a striking difference in the susceptibility of DHO-dehydrogenase/oxidase to Brequinar: apparent Ki = 6.09 +/- 0.05 (SD) nM (DHO; ubiquinone n = 10), but Ki = 3.10 +/- 0.09 (SD) mM (DHO; O2). Analyses of initial velocity experiments showed non-competitive inhibition of Brequinar with respect to the substrate dihydroorotic acid in both assays (dehydrogenase and oxidase). The inhibitory effect of the latter was compared to that of the competitive inhibitor 5-aza-dihydroorotate (apparent Ki = 15 +/- 0.25 (SD) microM). The present kinetic data on the action of the purified rodent DHO-DH with Brequinar and computer-aided analyses provide a better insight into the drug-enzyme interaction.


Assuntos
Compostos de Bifenilo/farmacologia , Di-Hidrorotato Oxidase/antagonistas & inibidores , Animais , Di-Hidrorotato Oxidase/isolamento & purificação , Cinética , Mitocôndrias Hepáticas/enzimologia , Modelos Químicos , Modelos Moleculares , Ratos , Relação Estrutura-Atividade
15.
Biochem Int ; 24(5): 833-9, 1991 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1663740

RESUMO

Dihydroorotate dehydrogenase purified from mitochondria of Plasmodium berghei, a rodent malaria parasite, mediates production of superoxide radical during oxidation of dihydroorotate to orotate. Reduction of dichlorophenolindophenol or cytochrome c or nitroblue tetrazolium was significantly inhibited by superoxide dismutase or theonyltrifluoroacetone, a specific iron chelator of the enzyme. These results, together with the recent evidence of manganese-superoxide dismutase activity in malarial mitochondria [Ranz, A., and Meshnick, S.R. (1989) Exp. Parasitol. 69, 125-128], suggest that the production of superoxide radical may occur in vivo.


Assuntos
Di-Hidrorotato Oxidase/metabolismo , Plasmodium berghei/enzimologia , Plasmodium falciparum/enzimologia , Superóxidos/metabolismo , 2,6-Dicloroindofenol/metabolismo , Animais , Grupo dos Citocromos c/metabolismo , Di-Hidrorotato Oxidase/isolamento & purificação , Eletroforese em Gel de Poliacrilamida , Mitocôndrias/metabolismo , Nitroazul de Tetrazólio/metabolismo , Ácido Orótico/análogos & derivados , Ácido Orótico/metabolismo , Oxirredução , Superóxido Dismutase/farmacologia
16.
Biochemistry ; 30(7): 1934-9, 1991 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-1847078

RESUMO

Dihydroorotate dehydrogenase (DHODase) has been purified 400-fold from the rodent malaria parasite Plasmodium berghei to apparent homogeneity by Triton X-100 solubilization followed by anion-exchange, Cibacron Blue F3GA-agarose affinity, and gel filtration chromatography. The purified enzyme has a molecular mass of 52 +/- 2 kDa on sodium dodecyl sulfate-polyacrylamide gel electrophoresis and of 55 +/- 6 kDa by gel filtration chromatography, and it has a pI of 8.2. It is active in monomeric form, contains 2.022 mol of iron and 1.602 acid-labile sulfurs per mole of enzyme, and does not contain a flavin cofactor. The purified DHODase exhibits optimal activity at pH 8.0 in the presence of the ubiquinone coenzyme CoQ6, CoQ7, CoQ9, or CoQ10. The Km values for L-DHO and CoQ6 are 7.9 +/- 2.5 microM and 21.6 +/- 5.5 microM, respectively. The kcat values for both substrates are 11.44 min-1 and 11.70 min-1, respectively. The reaction product orotate and an orotate analogue, 5-fluoroorotate, are competitive inhibitors of the enzyme-catalyzed reaction with Ki values of 30.5 microM and 34.9 microM, respectively. The requirement of the long-chain ubiquinones for activity supports the hypothesis of the linkage of pyrimidine biosynthesis to the electron transport system and oxygen utilization in malaria by DHODase via ubiquinones [Gutteridge, W. E., Dave, D., & Richards, W. H. G. (1979) Biochim. Biophys. Acta 582, 390-401].


Assuntos
Di-Hidrorotato Oxidase/isolamento & purificação , Plasmodium berghei/enzimologia , Animais , Western Blotting , Membrana Celular/enzimologia , Cromatografia de Afinidade , Cromatografia em Gel , Detergentes , Di-Hidrorotato Oxidase/metabolismo , Ferro/análise , Cinética , Peso Molecular , Octoxinol , Polietilenoglicóis , Enxofre/análise
17.
Biochem Int ; 22(6): 939-49, 1990 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1965281

RESUMO

Exposure of mouse L1210 leukemia cells to 25 microM brequinar for 4 h results in large accumulations of N-carbamyl-L-aspartate and L-dihydroorotate to cellular concentrations of 8.5 mM and 0.8 mM, respectively, while UTP and CTP decrease to 4% of their initial levels; incorporation of [14C]bicarbonate into nucleic acids (DNA and RNA) was decreased to 47%. These data provide direct evidence for inhibition of DHO dehydrogenase by brequinar in growing cells. Exposure of leukemia cells to 200 microM ciprofloxacin for 4 h did not affect de novo pyrimidine nucleotide biosynthesis or the incorporation of [14C]bicarbonate into nucleic acids but resulted in a general decrease in nucleoside triphosphates, with concomitant accumulation of nucleoside mono- and diphosphates (the adenylate energy charge decreased from 0.89 to 0.69), consistent with inhibition of the electron transport chain or uncoupling of oxidative phosphorylation.


Assuntos
Compostos de Bifenilo/farmacologia , Ciprofloxacina/farmacologia , Di-Hidrorotato Oxidase/antagonistas & inibidores , Nucleotídeos/biossíntese , Animais , Divisão Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , DNA de Neoplasias/biossíntese , Indução Enzimática , Leucemia L1210 , Camundongos , RNA Neoplásico/biossíntese , Células Tumorais Cultivadas
18.
J Biol Chem ; 265(24): 14242-9, 1990 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-2167311

RESUMO

Fluorinated compounds play an important role in enzymology as well as clinical medicine. Based on the stereochemical preferences of dihydroorotate oxidase and enzymes that use fluoroaspartate, it was anticipated that threo-5-fluoro-L-dihydroorotate (t-FDHO) would have the properties of an antimetabolite. Thus, t-FDHO was synthesized via the reduction of 5-fluoroorotate using NADH and dihydroorotate dehydrogenase that was free of dihydroorotase. When the product was purified and studied by high field proton and carbon 13 NMR, the fluorine, the five carbons, and all the nonexchangeable protons were readily observed. Confirmation of threo configuration was obtained by examining the vicinal coupling constants between the substituents on carbon 5 and carbon 6 of the newly synthesized compound. Additionally, t-FDHO could be reoxidized to 5-fluoroorotate in the presence of dihydroorotate dehydrogenase and NAD+. Treatment of t-FDHO with dihydroorotase generated N-carbamyl-threo-3-fluoro-L-aspartate (CTF-ASP) which was also purified and characterized by NMR. The antiproliferative activity of t-FDHO was determined against a diploid human fibrosarcoma cell line (HT-1080). Fifty microM t-FDHO caused 50% inhibition of HT-1080 cell proliferation. During the 48-h toxicity study, extracellular t-FDHO underwent significant hydrolysis to CTF-ASP. Further extracellular degradation to fluoroaspartate was not seen. The antiproliferative activity of t-FDHO was not due to extracellular degradation since CTF-ASP itself was essentially nontoxic.


Assuntos
Antineoplásicos/síntese química , Ácido Orótico/análogos & derivados , Linhagem Celular , Cromatografia por Troca Iônica , Di-Hidrorotato Oxidase/metabolismo , Fibrossarcoma , Humanos , Indicadores e Reagentes , Espectroscopia de Ressonância Magnética , Ácido Orótico/síntese química , Ácido Orótico/isolamento & purificação , Ácido Orótico/farmacologia , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos
19.
Cancer Res ; 50(15): 4644-9, 1990 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-2164443

RESUMO

Little is known about the in vivo effects of inhibition of the mitochondrial pyrimidine de novo synthesis enzyme dihydroorotic acid dehydrogenase (DHO-DH). In mice a new inhibitor of DHO-DH, Brequinar sodium (DUP-785, NSC 368390) depleted the plasma uridine concentration to 40% within 2 h, followed by a small rebound after 7-9 days. The drug was subsequently evaluated in a Phase I clinical trial, during which it was possible to follow its biochemical effects in 24 patients (27 courses). In addition to the measurement of plasma uridine concentrations, we also measured in lymphocytes of 9 patients (10 courses) the duration of DHO-DH inhibition. Brequinar sodium was administered every 3 weeks as an i.v. infusion at dose levels of 15-2250 mg/m2. The biochemical effects were studied following the first administration of the drug. In sonicated extracts of lymphocytes from 7 healthy volunteers the activity of DHO-DH varied from 2.0 to 3.9 nmol/h per 10(6) cells, while in the lymphocytes of 9 patients obtained immediately before treatment this value was between 0.5 and 4.8 nmol/h per 10(6) cells. Within 15 min of drug administration DHO-DH activity was not detectable and was still low up to 1 week later. Duration of the inhibition appeared to be related to the extent of clinical toxicity, e.g., myelosuppression, nausea, vomiting, diarrhea, and mucositis. Severe lymphopenia was observed in patients receiving Brequinar sodium at the maximum tolerated dose. At dose levels of greater than or equal to 600 mg/m2, uridine depletion (40-85%) was observed between 6 h and 4 days, followed by a rebound of 160-350% after 4-7 days. The extent of the depletion and of the accompanying rebound of uridine levels and the extent and duration of DHO-DH inhibition in the individual patients could be partially associated with drug toxicity in these patients. This is the first report describing biological effects of DHO-DH inhibition in humans in relation to the degree and duration of inhibition of this enzyme.


Assuntos
Antineoplásicos/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Di-Hidrorotato Oxidase/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Oxirredutases/antagonistas & inibidores , Animais , Compostos de Bifenilo/farmacocinética , Compostos de Bifenilo/farmacologia , Relação Dose-Resposta a Droga , Humanos , Linfócitos/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Valores de Referência , Uridina/sangue
20.
J Bacteriol ; 172(6): 3208-13, 1990 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1971621

RESUMO

The pyrC and pyrD genes of Escherichia coli K-12 encode the pyrimidine biosynthetic enzymes dihydroorotase and dihydroorotate dehydrogenase, respectively. A highly conserved sequence in the promoter regions of these two genes is similar to the pur operator, which is the binding site for the purine repressor (PurR). In this study, we examined the role of PurR in the regulation of pyrC and pyrD expression. Our results show that pyrC and pyrD expression was repressed approximately twofold in cells grown in the presence of adenine [corrected] through a mechanism requiring PurR. A mutation, designated pyrCp926, which alters a 6-base-pair region within the conserved sequence in the pyrC promoter eliminated PurR-mediated repression of pyrC expression. This result indicates that PurR binds to the pyrC (and presumably to the pyrD) conserved sequence and inhibits transcriptional initiation. We also demonstrated that the pyrCp926 mutation had no effect on pyrimidine-mediated regulation of pyrC expression, indicating that pyrimidine and purine effectors act through independent mechanisms to control the expression of the pyrC and pyrD genes.


Assuntos
Amidoidrolases/genética , Di-Hidro-Orotase/genética , Di-Hidrorotato Oxidase/genética , Escherichia coli/genética , Regulação Enzimológica da Expressão Gênica , Oxirredutases/genética , Purinas/farmacologia , Nucleotídeos de Pirimidina/biossíntese , Proteínas Repressoras/farmacologia , Fatores de Transcrição/farmacologia , Sequência de Bases , Escherichia coli/metabolismo , Dados de Sequência Molecular , Mutação , Pirimidinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...