Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Biol ; 402(2): 445-59, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20673832

RESUMEN

Ornithine δ-aminotransferase (OAT) of the malaria parasite Plasmodium falciparum catalyzes the reversible conversion of ornithine into glutamate-5-semialdehyde and glutamate and is-in contrast to its human counterpart-activated by thioredoxin (Trx) by a factor of 10. Trx, glutaredoxin, and plasmoredoxin are redox-active proteins that play a crucial role in the maintenance and control of redox reactions, and were shown to interact with P. falciparum OAT. OAT, which is involved in ornithine homeostasis and proline biosynthesis, is essential for mitotic cell division in rapidly growing cells, thus representing a potential target for chemotherapeutic intervention. Here we report the three-dimensional crystal structure of P. falciparum OAT at 2.3 Å resolution. The overall structure is very similar to that of the human OAT. However, in plasmodial OAT, the loop involved in substrate binding contains two cysteine residues, which are lacking in human OAT. Site-directed mutagenesis of these cysteines and functional analysis demonstrated that Cys154 and Cys163 mediate the interaction with Trx. Interestingly, the Cys154→Ser mutant has a strongly reduced specific activity, most likely due to impaired binding of ornithine. Cys154 and Cys163 are highly conserved in Plasmodium but do not exist in other organisms, suggesting that redox regulation of OAT by Trx is specific for malaria parasites. Plasmodium might require a tight Trx-mediated control of OAT activity for coordinating ornithine homeostasis, polyamine synthesis, proline synthesis, and mitotic cell division.


Asunto(s)
Ornitina-Oxo-Ácido Transaminasa/química , Plasmodium falciparum/enzimología , Proteínas Protozoarias/química , Sustitución de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Cisteína/genética , Cisteína/metabolismo , Activadores de Enzimas/farmacología , Ácido Glutámico/metabolismo , Cinética , Redes y Vías Metabólicas , Modelos Biológicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Ornitina/metabolismo , Ornitina-Oxo-Ácido Transaminasa/metabolismo , Oxidación-Reducción , Plasmodium falciparum/metabolismo , Estructura Terciaria de Proteína , Proteínas Protozoarias/metabolismo , Tiorredoxinas/farmacología
2.
PLoS Pathog ; 6(12): e1001242, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21203490

RESUMEN

Malaria, caused by the apicomplexan parasite Plasmodium, still represents a major threat to human health and welfare and leads to about one million human deaths annually. Plasmodium is a rapidly multiplying unicellular organism undergoing a complex developmental cycle in man and mosquito - a life style that requires rapid adaptation to various environments. In order to deal with high fluxes of reactive oxygen species and maintain redox regulatory processes and pathogenicity, Plasmodium depends upon an adequate redox balance. By systematically studying the subcellular localization of the major antioxidant and redox regulatory proteins, we obtained the first complete map of redox compartmentation in Plasmodium falciparum. We demonstrate the targeting of two plasmodial peroxiredoxins and a putative glyoxalase system to the apicoplast, a non-photosynthetic plastid. We furthermore obtained a complete picture of the compartmentation of thioredoxin- and glutaredoxin-like proteins. Notably, for the two major antioxidant redox-enzymes--glutathione reductase and thioredoxin reductase--Plasmodium makes use of alternative-translation-initiation (ATI) to achieve differential targeting. Dual localization of proteins effected by ATI is likely to occur also in other Apicomplexa and might open new avenues for therapeutic intervention.


Asunto(s)
Compartimento Celular/fisiología , Peroxirredoxinas/metabolismo , Plasmodium falciparum/metabolismo , Glutatión Reductasa/metabolismo , Oxidación-Reducción , Plasmodium falciparum/citología , Plasmodium falciparum/enzimología , Plastidios/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo
3.
PLoS Pathog ; 5(4): e1000383, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19360125

RESUMEN

The malarial parasite Plasmodium falciparum possesses a functional thioredoxin and glutathione system comprising the dithiol-containing redox proteins thioredoxin (Trx) and glutaredoxin (Grx), as well as plasmoredoxin (Plrx), which is exclusively found in Plasmodium species. All three proteins belong to the thioredoxin superfamily and share a conserved Cys-X-X-Cys motif at the active site. Only a few of their target proteins, which are likely to be involved in redox reactions, are currently known. The aim of the present study was to extend our knowledge of the Trx-, Grx-, and Plrx-interactome in Plasmodium. Based on the reaction mechanism, we generated active site mutants of Trx and Grx lacking the resolving cysteine residue. These mutants were bound to affinity columns to trap target proteins from P. falciparum cell extracts after formation of intermolecular disulfide bonds. Covalently linked proteins were eluted with dithiothreitol and analyzed by mass spectrometry. For Trx and Grx, we were able to isolate 17 putatively redox-regulated proteins each. Furthermore, the approach was successfully established for Plrx, leading to the identification of 21 potential target proteins. In addition to confirming known interaction partners, we captured potential target proteins involved in various processes including protein biosynthesis, energy metabolism, and signal transduction. The identification of three enzymes involved in S-adenosylmethionine (SAM) metabolism furthermore suggests that redox control is required to balance the metabolic fluxes of SAM between methyl-group transfer reactions and polyamine synthesis. To substantiate our data, the binding of the redoxins to S-adenosyl-L-homocysteine hydrolase and ornithine aminotransferase (OAT) were verified using BIAcore surface plasmon resonance. In enzymatic assays, Trx was furthermore shown to enhance the activity of OAT. Our approach led to the discovery of several putatively redox-regulated proteins, thereby contributing to our understanding of the redox interactome in malarial parasites.


Asunto(s)
Plasmodium falciparum/química , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Tiorredoxinas/química , Tiorredoxinas/metabolismo , Animales , Electroforesis en Gel Bidimensional , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Reacción en Cadena de la Polimerasa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Resonancia por Plasmón de Superficie
4.
Antimicrob Agents Chemother ; 53(2): 622-30, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19015351

RESUMEN

The cancer chemopreventive agent ellagic acid (EA) is a known inhibitor of glutathione S-transferases (GSTs) and possesses antiplasmodial activities in the upper-nanomolar range. In the recent drug development approach, the properties of the active site of Plasmodium falciparum GST were exploited for inhibitor design by introducing one or two additional hydroxyl groups into EA, yielding flavellagic acid (FEA) and coruleoellagic acid (CEA), respectively. Indeed, the inhibition of P. falciparum GST was improved with the increasing hydrophilicity of the planar polyaromatic ring system. Studying the effects of the two compounds on the central redox enzymes of Plasmodium revealed that glutathione reductase and thioredoxin reductase also are inhibited in the lower-micromolar range. Both compounds had strong antiplasmodial activity in the lower-nanomolar range and were particularly effective against chloroquine (CQ)-resistant P. falciparum strains. Neither FEA nor CEA showed cytotoxic effects on human cells. This was supported by negligible changes in transcript levels and enzyme activities of redox enzymes in human A549 cells upon treatment with the compounds. In Plasmodium, however, CEA treatment resulted in a marked downregulation of most antioxidant genes studied and impaired mainly the trophozoite stage of the parasites. In addition, EA, CEA, and FEA were found to strongly inhibit in vitro heme aggregation. In vitro and preliminary in vivo studies indicated that, compared to CQ, CEA is a slowly acting compound and is able to significantly improve the survival of Plasmodium berghei-infected mice. We conclude that FEA and CEA are promising antimalarial compounds that deserve to be studied further.


Asunto(s)
Antimaláricos/farmacología , Ácido Elágico/análogos & derivados , Ácido Elágico/farmacología , Plasmodium falciparum/efectos de los fármacos , Animales , Antimaláricos/química , Células Cultivadas , Cloroquina/farmacología , ADN Complementario/biosíntesis , ADN Complementario/genética , Resistencia a Medicamentos , Ácido Elágico/química , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Hemo/química , Humanos , Cinética , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Biol Chem ; 282(39): 28419-28430, 2007 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-17664277

RESUMEN

Glyoxalase I (GloI) catalyzes the glutathione-dependent conversion of 2-oxoaldehydes to S-2-hydroxyacylglutathione derivatives. Studies on GloI from diverse organisms such as man, bacteria, yeast, and different parasites show striking differences among these potentially isofunctional enzymes as far as metal content and the number of active sites per subunit are concerned. So far, it is not known whether this structural variability is linked to catalytic or regulatory features in vivo. Here we show that recombinant GloI from the malaria parasite Plasmodium falciparum has a high- and a low-affinity binding site for the diastereomeric hemithioacetals formed by addition of glutathione to methylglyoxal. Both active sites of the monomeric enzyme are functional and have similar k(cat)(app) values. Proteolytic susceptibility studies and detailed analyses of the steady-state kinetics of active-site mutants suggest that both reaction centers can adopt two discrete conformations and are allosterically coupled. As a result of the positive homotropic allosteric coupling, P. falciparum GloI has an increased affinity at low substrate concentrations and an increased activity at higher substrate concentrations. This could also be the case for GloI from yeast and other organisms. Potential physiologically relevant differences between monomeric GloI and homodimeric GloI are discussed. Our results provide a strong basis for drug development strategies and significantly enhance our understanding of GloI kinetics and structure-function relationships. Furthermore, they extend the current knowledge on allosteric regulation of monomeric proteins in general.


Asunto(s)
Lactoilglutatión Liasa/química , Plasmodium falciparum/enzimología , Proteínas Protozoarias/química , Regulación Alostérica/fisiología , Animales , Bacterias/enzimología , Sitios de Unión/fisiología , Diseño de Fármacos , Inhibidores Enzimáticos/uso terapéutico , Glutatión/química , Glutatión/metabolismo , Humanos , Cinética , Lactoilglutatión Liasa/antagonistas & inhibidores , Lactoilglutatión Liasa/metabolismo , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/enzimología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Piruvaldehído/química , Piruvaldehído/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad de la Especie , Relación Estructura-Actividad , Levaduras/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...