Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Antimicrob Agents Chemother ; 68(5): e0169023, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38501806

RESUMEN

Malaria tropica, caused by the parasite Plasmodium falciparum (P. falciparum), remains one of the greatest public health burdens for humankind. Due to its pivotal role in parasite survival, the energy metabolism of P. falciparum is an interesting target for drug design. To this end, analysis of the central metabolite adenosine triphosphate (ATP) is of great interest. So far, only cell-disruptive or intensiometric ATP assays have been available in this system, with various drawbacks for mechanistic interpretation and partly inconsistent results. To address this, we have established fluorescent probes, based on Förster resonance energy transfer (FRET) and known as ATeam, for use in blood-stage parasites. ATeams are capable of measuring MgATP2- levels in a ratiometric manner, thereby facilitating in cellulo measurements of ATP dynamics in real-time using fluorescence microscopy and plate reader detection and overcoming many of the obstacles of established ATP analysis methods. Additionally, we established a superfolder variant of the ratiometric pH sensor pHluorin (sfpHluorin) in P. falciparum to monitor pH homeostasis and control for pH fluctuations, which may affect ATeam measurements. We characterized recombinant ATeam and sfpHluorin protein in vitro and stably integrated the sensors into the genome of the P. falciparum NF54attB cell line. Using these new tools, we found distinct sensor response patterns caused by several different drug classes. Arylamino alcohols increased and redox cyclers decreased ATP; doxycycline caused first-cycle cytosol alkalization; and 4-aminoquinolines caused aberrant proteolysis. Our results open up a completely new perspective on drugs' mode of action, with possible implications for target identification and drug development.


Asunto(s)
Adenosina Trifosfato , Antimaláricos , Transferencia Resonante de Energía de Fluorescencia , Plasmodium falciparum , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Plasmodium falciparum/genética , Adenosina Trifosfato/metabolismo , Antimaláricos/farmacología , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes/química , Humanos , Quinina/farmacología , Doxiciclina/farmacología , Artemisininas/farmacología , Cloroquina/farmacología , Concentración de Iones de Hidrógeno
2.
Int J Mol Sci ; 24(16)2023 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-37628920

RESUMEN

The protozoan parasite Plasmodium falciparum is the causative pathogen of the most severe form of malaria, for which novel strategies for treatment are urgently required. The primary energy supply for intraerythrocytic stages of Plasmodium is the production of ATP via glycolysis. Due to the parasite's strong dependence on this pathway and the significant structural differences of its glycolytic enzymes compared to its human counterpart, glycolysis is considered a potential drug target. In this study, we provide the first three-dimensional protein structure of P. falciparum hexokinase (PfHK) containing novel information about the mechanisms of PfHK. We identified for the first time a Plasmodium-specific insertion that lines the active site. Moreover, we propose that this insertion plays a role in ATP binding. Residues of the insertion further seem to affect the tetrameric interface and therefore suggest a special way of communication among the different monomers. In addition, we confirmed that PfHK is targeted and affected by oxidative posttranslational modifications (oxPTMs). Both S-glutathionylation and S-nitrosation revealed an inhibitory effect on the enzymatic activity of PfHK.


Asunto(s)
Malaria Falciparum , Plasmodium , Humanos , Plasmodium falciparum , Hexoquinasa , Catálisis , Adenosina Trifosfato
3.
Int J Mol Sci ; 24(10)2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37239962

RESUMEN

As unicellular parasites are highly dependent on NADPH as a source for reducing equivalents, the main NADPH-producing enzymes glucose 6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD) of the pentose phosphate pathway are considered promising antitrypanosomatid drug targets. Here we present the biochemical characterization and crystal structure of Leishmania donovani 6PGD (Ld6PGD) in complex with NADP(H). Most interestingly, a previously unknown conformation of NADPH is visible in this structure. In addition, we identified auranofin and other gold(I)-containing compounds as efficient Ld6PGD inhibitors, although it has so far been assumed that trypanothione reductase is the sole target of auranofin in Kinetoplastida. Interestingly, 6PGD from Plasmodium falciparum is also inhibited at lower micromolar concentrations, whereas human 6PGD is not. Mode-of-inhibition studies indicate that auranofin competes with 6PG for its binding site followed by a rapid irreversible inhibition. By analogy with other enzymes, this suggests that the gold moiety is responsible for the observed inhibition. Taken together, we identified gold(I)-containing compounds as an interesting class of inhibitors against 6PGDs from Leishmania and possibly from other protozoan parasites. Together with the three-dimensional crystal structure, this provides a valid basis for further drug discovery approaches.


Asunto(s)
Leishmania donovani , Leishmaniasis , Humanos , Leishmania donovani/metabolismo , Oro/farmacología , Auranofina/farmacología , Fosfogluconato Deshidrogenasa/química , Fosfogluconato Deshidrogenasa/metabolismo , NADP/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo
4.
Eur J Med Chem ; 251: 115179, 2023 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-36948075

RESUMEN

Schistosomiasis is an infectious disease caused by blood flukes of the genus Schistosoma and affects approximately 200 million people worldwide. Since Praziquantel (PZQ) is the only drug for schistosomiasis, alternatives are needed. By a biochemical approach, we identified a tegumentally expressed aldehyde dehydrogenase (ALDH) of S. mansoni, SmALDH_312. Molecular analyses of adult parasites showed Smaldh_312 transcripts in both genders and different tissues. Physiological and cell-biological experiments exhibited detrimental effects of the drug disulfiram (DSF), a known ALDH inhibitor, on larval and adult schistosomes in vitro. DSF also reduced stem-cell proliferation and caused severe tegument damage in treated worms. In silico-modelling of SmALDH_312 and docking analyses predicted DSF binding, which we finally confirmed by enzyme assays with recombinant SmALDH_312. Furthermore, we identified compounds of the Medicine for Malaria Venture (MMV) pathogen box inhibiting SmALDH_312 activity. Our findings represent a promising starting point for further development towards new drugs for schistosomiasis.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomiasis , Animales , Femenino , Masculino , Schistosoma mansoni , Esquistosomiasis mansoni/tratamiento farmacológico , Disulfiram/farmacología , Disulfiram/uso terapéutico , Aldehído Deshidrogenasa/farmacología
5.
Commun Biol ; 5(1): 1353, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494598

RESUMEN

Since unicellular parasites highly depend on NADPH as a source for reducing equivalents, the pentose phosphate pathway, especially the first and rate-limiting NADPH-producing enzyme glucose 6-phosphate dehydrogenase (G6PD), is considered an excellent antitrypanosomatid drug target. Here we present the crystal structure of Leishmania donovani G6PD (LdG6PD) elucidating the unique N-terminal domain of Kinetoplastida G6PDs. Our investigations on the function of the N-domain suggest its involvement in the formation of a tetramer that is completely different from related Trypanosoma G6PDs. Structural and functional investigations further provide interesting insights into the binding mode of LdG6PD, following an ordered mechanism, which is confirmed by a G6P-induced domain shift and rotation of the helical N-domain. Taken together, these insights into LdG6PD contribute to the understanding of G6PDs' molecular mechanisms and provide an excellent basis for further drug discovery approaches.


Asunto(s)
Leishmania donovani , Leishmania donovani/genética , NADP/química , NADP/metabolismo , Vía de Pentosa Fosfato , Glucosa , Fosfatos
6.
Structure ; 30(10): 1452-1461.e3, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35998635

RESUMEN

The protozoan parasite Plasmodium falciparum causes the most severe form of malaria and is highly dependent on glycolysis. Glycolytic enzymes were shown to be massively redox regulated, inter alia via oxidative post-translational modifications (oxPTMs) of their cysteine residues. In this study, we identified P. falciparum pyruvate kinase (PfPK) C49 and C343 as amino acid residues essentially involved in maintaining structural and functional integrity of the enzyme. The mutation of these cysteines resulted in an altered substrate affinity, lower enzymatic activities, and, as studied by X-ray crystallography, conformational changes within the A-domain where the substrate binding site is located. Although the loss of a cysteine evoked an impaired catalysis in both mutants, the effects observed for mutant C49A were more severe: multiple conformational changes, caused by the loss of two hydrogen bonds, impeded proper substrate binding and thus the transfer of phosphate upon catalysis.


Asunto(s)
Cisteína , Plasmodium falciparum , Cisteína/metabolismo , Glucólisis , Fosfatos/metabolismo , Proteínas Protozoarias/química , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo
7.
Cell Rep ; 39(11): 110923, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705035

RESUMEN

The uptake and digestion of host hemoglobin by malaria parasites during blood-stage growth leads to significant oxidative damage of membrane lipids. Repair of lipid peroxidation damage is crucial for parasite survival. Here, we demonstrate that Plasmodium falciparum imports a host antioxidant enzyme, peroxiredoxin 6 (PRDX6), during hemoglobin uptake from the red blood cell cytosol. PRDX6 is a lipid-peroxidation repair enzyme with phospholipase A2 (PLA2) activity. Inhibition of PRDX6 with a PLA2 inhibitor, Darapladib, increases lipid-peroxidation damage in the parasite and disrupts transport of hemoglobin-containing vesicles to the food vacuole, causing parasite death. Furthermore, inhibition of PRDX6 synergistically reduces the survival of artemisinin-resistant parasites following co-treatment of parasite cultures with artemisinin and Darapladib. Thus, PRDX6 is a host-derived drug target for development of antimalarial drugs that could help overcome artemisinin resistance.


Asunto(s)
Antimaláricos , Artemisininas , Malaria Falciparum , Malaria , Peroxiredoxina VI , Animales , Antimaláricos/farmacología , Artemisininas/metabolismo , Artemisininas/farmacología , Benzaldehídos/farmacología , Resistencia a Medicamentos , Hemoglobinas/metabolismo , Humanos , Lípidos , Malaria/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Ratones , Oximas/farmacología , Peroxiredoxina VI/inmunología , Peroxiredoxina VI/metabolismo , Plasmodium falciparum
8.
Curr Res Struct Biol ; 4: 87-95, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35434650

RESUMEN

Plasmoredoxin is a 22 â€‹kDa thiol-disulfide oxidoreductase involved in cellular redox regulatory processes and antioxidant defense. The 1.6 â€‹Å structure of the protein, solved via X-ray crystallography, adopts a modified thioredoxin fold. The structure reveals that plasmoredoxin, unique for malarial parasites, forms a new subgroup of thioredoxin-like proteins together with tryparedoxin, unique for kinetoplastids. Unlike most members of this superfamily, Plrx does not have a proline residue within the CxxC redox motif. In addition, the Plrx structure has a distinct C-terminal domain. Similar to human thioredoxin, plasmoredoxin forms monomers and dimers, which are also structurally similar to the human thioredoxin dimer, and, as in humans, plasmoredoxin is inactive as a dimer. Monomer-dimer equilibrium depends on the surrounding redox conditions, which could support the parasite in reacting to oxidative challenges. Based on structural considerations, the residues of the dimer interface are likely to interact with target proteins. In contrast to human and Plasmodium falciparum thioredoxin, however, there is a cluster of positively charged residues at the dimer interface of plasmoredoxin. These intersubunit (lysine) residues might allow binding of the protein to cellular membranes or to plasminogen. Malaria parasites lack catalase and glutathione peroxidase and therefore depend on their other glutathione and thioredoxin-dependent redox relays. Plasmoredoxin could be part of a so far unknown electron transfer system that only occurs in these parasites. Since the surface charge of plasmoredoxin differs significantly from other members of the thioredoxin superfamily, its three-dimensional structure can provide a model for designing selective redox-modulatory inhibitors.

9.
Antimicrob Agents Chemother ; 66(4): e0210921, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35266827

RESUMEN

In Plasmodium, the first two and rate-limiting enzymes of the pentose phosphate pathway, glucose 6-phosphate dehydrogenase (G6PD) and the 6-phosphogluconolactonase, are bifunctionally fused to a unique enzyme named GluPho, differing structurally and mechanistically from the respective human orthologs. Consistent with the enzyme's essentiality for malaria parasite proliferation and propagation, human G6PD deficiency has immense impact on protection against severe malaria, making PfGluPho an attractive antimalarial drug target. Herein we report on the optimized lead compound N-(((2R,4S)-1-cyclobutyl-4-hydroxypyrrolidin-2-yl)methyl)-6-fluoro-4-methyl-11-oxo-10,11-dihydrodibenzo[b,f][1,4]thiazepine-8-carboxamide (SBI-0797750), a potent and fully selective PfGluPho inhibitor with robust nanomolar activity against recombinant PfGluPho, PvG6PD, and P. falciparum blood-stage parasites. Mode-of-action studies have confirmed that SBI-0797750 disturbs the cytosolic glutathione-dependent redox potential, as well as the cytosolic and mitochondrial H2O2 homeostasis of P. falciparum blood stages, at low nanomolar concentrations. Moreover, SBI-0797750 does not harm red blood cell (RBC) integrity and phagocytosis and thus does not promote anemia. SBI-0797750 is therefore a very promising antimalarial lead compound.


Asunto(s)
Antimaláricos , Deficiencia de Glucosafosfato Deshidrogenasa , Malaria Falciparum , Malaria Vivax , Malaria , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Hidrolasas de Éster Carboxílico , Glucosa/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Malaria Falciparum/tratamiento farmacológico , Malaria Vivax/tratamiento farmacológico , Fosfatos , Plasmodium falciparum/metabolismo , Plasmodium vivax
10.
Antioxid Redox Signal ; 37(1-3): 1-18, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35072524

RESUMEN

Aims: Genetically encoded green fluorescent protein (GFP)-based redox biosensors are widely used to monitor specific and dynamic redox processes in living cells. Over the last few years, various biosensors for a variety of applications were engineered and enhanced to match the organism and cellular environments, which should be investigated. In this context, the unicellular intraerythrocytic parasite Plasmodium, the causative agent of malaria, represents a challenge, as the small size of the organism results in weak fluorescence signals that complicate precise measurements, especially for cell compartment-specific observations. To address this, we have functionally and structurally characterized an enhanced redox biosensor superfolder roGFP2 (sfroGFP2). Results: SfroGFP2 retains roGFP2-like behavior, yet with improved fluorescence intensity (FI) in cellulo. SfroGFP2-based redox biosensors are pH insensitive in a physiological pH range and show midpoint potentials comparable with roGFP2-based redox biosensors. Using crystallography and rigidity theory, we identified the superfolding mutations as being responsible for improved structural stability of the biosensor in a redox-sensitive environment, thus explaining the improved FI in cellulo. Innovation: This work provides insight into the structure and function of GFP-based redox biosensors. It describes an improved redox biosensor (sfroGFP2) suitable for measuring oxidizing effects within small cells where applicability of other redox sensor variants is limited. Conclusion: Improved structural stability of sfroGFP2 gives rise to increased FI in cellulo. Fusion to hGrx1 (human glutaredoxin-1) provides the hitherto most suitable biosensor for measuring oxidizing effects in Plasmodium. This sensor is of major interest for studying glutathione redox changes in small cells, as well as subcellular compartments in general. Antioxid. Redox Signal. 37, 1-18.


Asunto(s)
Técnicas Biosensibles , Glutatión , Plasmodium , Técnicas Biosensibles/métodos , Glutatión/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Oxidación-Reducción , Plasmodium/aislamiento & purificación
11.
Parasitol Int ; 87: 102513, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34785370

RESUMEN

Plasmodium falciparum macrophage migration inhibitory factor (PfMIF) is a homologue of the multifunctional human host cytokine MIF (HsMIF). Upon schizont rupture it is released into the human blood stream where it acts as a virulence factor, modulating the host immune system. Whereas for HsMIF a tautomerase, an oxidoreductase, and a nuclease activity have been identified, the latter has not yet been studied for PfMIF. Furthermore, previous studies identified PfMIF as a target for several redox post-translational modifications. Therefore, we analysed the impact of S-glutathionylation and S-nitrosation on the protein's functions. To determine the impact of the four cysteines of PfMIF we produced His-tagged cysteine to alanine mutants of PfMIF via site-directed mutagenesis. Recombinant proteins were analysed via mass spectrometry, and enzymatic assays. Here we show for the first time that PfMIF acts as a DNase of human genomic DNA and that this activity is greater than that shown by HsMIF. Moreover, we observed a significant decrease in the maximum velocity of the DCME tautomerase activity of PfMIF upon alanine replacement of Cys3, and Cys3/Cys4 double mutant. Lastly, using a yeast reporter system, we were able to verify binding of PfMIF to the human chemokine receptors CXCR4, and demonstrate a so-far overlooked binding to CXCR2, both of which function as non-cognate receptors for HsMIF. While S-glutathionylation and S-nitrosation of PfMIF did not impair the tautomerase activity of PfMIF, activation of these receptors was significantly decreased.


Asunto(s)
Cisteína/deficiencia , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/genética , Plasmodium falciparum/química , Alanina/química , Cisteína/genética , Desoxirribonucleasas/metabolismo , Humanos , Plasmodium falciparum/genética , Proteínas Recombinantes/genética
12.
Sci Rep ; 9(1): 13542, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31537845

RESUMEN

Peroxiredoxins (Prxs) are crucially involved in maintaining intracellular H2O2 homeostasis via their peroxidase activity. However, more recently, this class of proteins was found to also transmit oxidizing equivalents to selected downstream proteins, which suggests an important function of Prxs in the regulation of cellular protein redox relays. Using a pull-down assay based on mixed disulfide fishing, we characterized the thiol-dependent interactome of cytosolic Prx1a and mitochondrial Prx1m from the apicomplexan malaria parasite Plasmodium falciparum (Pf). Here, 127 cytosolic and 20 mitochondrial proteins that are components of essential cellular processes were found to interact with PfPrx1a and PfPrx1m, respectively. Notably, our data obtained with active-site mutants suggests that reducing equivalents might also be transferred from Prxs to target proteins. Initial functional analyses indicated that the interaction with Prx can strongly impact the activity of target proteins. The results provide initial insights into the interactome of Prxs at the level of a eukaryotic whole cell proteome. Furthermore, they contribute to our understanding of redox regulatory principles and thiol-dependent redox relays of Prxs in subcellular compartments.


Asunto(s)
Peroxirredoxinas/metabolismo , Plasmodium falciparum/metabolismo , Antioxidantes/metabolismo , Cisteína/metabolismo , Citosol/metabolismo , Disulfuros/metabolismo , Peróxido de Hidrógeno/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Peroxirredoxinas/fisiología , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Compuestos de Sulfhidrilo
14.
Malar J ; 18(1): 22, 2019 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683097

RESUMEN

BACKGROUND: Since malaria parasites highly depend on ribose 5-phosphate for DNA and RNA synthesis and on NADPH as a source of reducing equivalents, the pentose phosphate pathway (PPP) is considered an excellent anti-malarial drug target. In Plasmodium, a bifunctional enzyme named glucose 6-phosphate dehydrogenase 6-phosphogluconolactonase (GluPho) catalyzes the first two steps of the PPP. PfGluPho has been shown to be essential for the growth of blood stage Plasmodium falciparum parasites. METHODS: Plasmodium vivax glucose 6-phosphate dehydrogenase (PvG6PD) was cloned, recombinantly produced in Escherichia coli, purified, and characterized via enzyme kinetics and inhibitor studies. The effects of post-translational cysteine modifications were assessed via western blotting and enzyme activity assays. Genetically encoded probes were employed to study the effects of G6PD inhibitors on the cytosolic redox potential of Plasmodium. RESULTS: Here the recombinant production and characterization of PvG6PD, the C-terminal and NADPH-producing part of PvGluPho, is described. A comparison with PfG6PD (the NADPH-producing part of PfGluPho) indicates that the P. vivax enzyme has higher KM values for the substrate and cofactor. Like the P. falciparum enzyme, PvG6PD is hardly affected by S-glutathionylation and moderately by S-nitrosation. Since there are several naturally occurring variants of PfGluPho, the impact of these mutations on the kinetic properties of the enzyme was analysed. Notably, in contrast to many human G6PD variants, the mutations resulted in only minor changes in enzyme activity. Moreover, nanomolar IC50 values of several compounds were determined on P. vivax G6PD (including ellagic acid, flavellagic acid, and coruleoellagic acid), inhibitors that had been previously characterized on PfGluPho. ML304, a recently developed PfGluPho inhibitor, was verified to also be active on PvG6PD. Using genetically encoded probes, ML304 was confirmed to disturb the cytosolic glutathione-dependent redox potential of P. falciparum blood stage parasites. Finally, a new series of novel small molecules with the potential to inhibit the falciparum and vivax enzymes were synthesized, resulting in two compounds with nanomolar activity. CONCLUSION: The characterization of PvG6PD makes this enzyme accessible to further drug discovery activities. In contrast to naturally occurring G6PD variants in the human host that can alter the kinetic properties of the enzyme and thus the redox homeostasis of the cells, the naturally occurring PfGluPho variants studied here are unlikely to have a major impact on the parasites' redox homeostasis. Several classes of inhibitors have been successfully tested and are presently being followed up.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Glucosafosfato Deshidrogenasa/genética , Malaria Vivax/genética , Complejos Multienzimáticos/genética , Proteínas Protozoarias/genética , Hidrolasas de Éster Carboxílico/metabolismo , Clonación Molecular , Citosol/metabolismo , Escherichia coli/metabolismo , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Glucosafosfato Deshidrogenasa/metabolismo , Cinética , Malaria Vivax/enzimología , Malaria Vivax/metabolismo , Complejos Multienzimáticos/metabolismo , Oxidación-Reducción , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
15.
ACS Infect Dis ; 4(11): 1601-1612, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30129748

RESUMEN

Studying redox metabolism in malaria parasites is of great interest for understanding parasite biology, parasite-host interactions, and mechanisms of drug action. Genetically encoded fluorescent redox sensors have recently been described as powerful tools for determining the glutathione-dependent redox potential in living parasites. In the present study, we genomically integrated and expressed the ratiometric redox sensors hGrx1-roGFP2 (human glutaredoxin 1 fused to reduction-oxidation sensitive green fluorescent protein) and sfroGFP2 (superfolder roGFP2) in the cytosol of NF54- attB blood-stage Plasmodium falciparum parasites. Both sensors were evaluated in vitro and in cell culture with regard to their fluorescence properties and reactivity. As genomic integration allows for the stable expression of redox sensors in parasites, we systematically compared single live-cell imaging with plate reader detection. For these comparisons, short-term effects of redox-active compounds were analyzed along with mid- and long-term effects of selected antimalarial agents. Of note, the single components of the redox probes themselves did not influence the redox balance of the parasites. Our analyses revealed comparable results for both the hGrx1-roGFP2 and sfroGFP2 probes, with sfroGFP2 exhibiting a more pronounced fluorescence intensity in cellulo. Accordingly, the sfroGFP2 probe was employed to monitor the fluorescence signals throughout the parasites' asexual life cycle. Through the use of stable genomic integration, we demonstrate a means of overcoming the limitations of transient transfection, allowing more detailed in-cell studies as well as high-throughput analyses using plate reader-based approaches.


Asunto(s)
Colorantes Fluorescentes , Glutarredoxinas/análisis , Interacciones Huésped-Parásitos , Plasmodium falciparum/metabolismo , Antimaláricos/farmacología , Citosol/efectos de los fármacos , Citosol/parasitología , Fluorescencia , Proteínas Fluorescentes Verdes/análisis , Humanos , Oxidación-Reducción , Plasmodium falciparum/efectos de los fármacos , Proteínas Recombinantes/análisis , Transfección
16.
J Mol Biol ; 430(21): 4049-4067, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30098336

RESUMEN

The enzyme 6-phosphogluconate dehydrogenase (6PGD) of the malaria parasite Plasmodium falciparum catalyzes the third step of the pentose phosphate pathway converting 6-phosphogluconate (6PG) to ribulose 5-phosphate. The NADPH produced by 6PGD is crucial for antioxidant defense and redox regulation, and ribose 5-phosphate is essential for DNA and RNA synthesis in the rapidly growing parasite. Thus, 6PGD represents an attractive antimalarial drug target. In this study, we present the X-ray structures of Pf6PGD in native form as well as in complex with 6PG or nicotinamide adenine dinucleotide phosphate (NADP+) at resolutions of 2.8, 1.9, and 2.9 Å, respectively. The overall structure of the protein is similar to structures of 6PGDs from other species; however, a flexible loop close to the active site rearranges upon binding of 6PG and likely regulates the conformation of the cofactor NADP+. Upon binding of 6PG, the active site loop adopts a closed conformation. In the absence of 6PG, the loop opens and NADP+ is bound in a waiting position, indicating that the cofactor and 6PG bind independently from each other. This sequential binding mechanism was supported by kinetic studies on the homodimeric wild-type Pf6PGD. Furthermore, the function of the Plasmodium-specific residue W104L mutant was characterized by site-directed mutagenesis. Notably, the activity of Pf6PGD was found to be post-translationally redox regulated via S-nitrosylation, and screening the Medicines for Malaria Venture Malaria Box identified several compounds with IC50s in the low micromolar range. Together with the three-dimensional structure of the protein, this is a promising starting point for further drug discovery approaches.


Asunto(s)
Antimaláricos/química , Inhibidores Enzimáticos/química , Fosfogluconato Deshidrogenasa/química , Plasmodium falciparum/enzimología , Secuencia de Aminoácidos , Antimaláricos/farmacología , Sitios de Unión , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Fenómenos Mecánicos , Modelos Moleculares , Conformación Molecular , Fosfogluconato Deshidrogenasa/antagonistas & inhibidores , Fosfogluconato Deshidrogenasa/metabolismo , Plasmodium falciparum/efectos de los fármacos , Unión Proteica , Proteínas Recombinantes , Relación Estructura-Actividad , Especificidad por Sustrato
17.
FEBS J ; 285(3): 542-558, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29222842

RESUMEN

In most cells, the thioredoxin (Trx) and glutathione systems are essential in maintaining redox homeostasis. The selenoprotein thioredoxin glutathione reductase (TGR) is a hybrid enzyme in which a glutaredoxin (Grx) domain is linked to a thioredoxin reductase (TrxR). Notably, the protein is also capable of reducing glutathione disulfide (GSSG), thus representing an important link between the two redox systems. In this study, we recombinantly produced human TGR (hTGR wild-type) by fusing its open reading frame with a bacterial selenocysteine insertion sequence element and co-expressing the construct in Escherichia coli together with the selA, selB, and selC genes. Additionally, the Sec→Cys mutant (hTGRU642C ) of the full-length protein, the isolated TrxR domain (hTGR151-643 ) and the Grx domain containing a monothiol active site (hTGR1-150 ) were produced and purified. All four proteins were kinetically characterized in direct comparison using Trx, DTNB, HED, or GSSG as the oxidizing substrate. Interestingly, the HED reduction activity was Sec independent and comparable in the full-length protein and the isolated Grx domain, whereas the TrxR and glutathione reductase reactions were clearly selenocysteine dependent, with the GR reaction requiring the Grx domain. Site-directed mutagenesis studies revealed novel insights into the mechanism of GSSG reduction. Furthermore, we identified several glutathionylation sites in hTGR, including Cys93, Cys133, and Cys619, and an inhibitory effect of these modifications on enzyme activity. In contrast to other TGRs, for example, from platyhelminth parasites, hTGR did not exhibit hysteretic behavior. These findings provide new insights into the reaction mechanism and regulation of monothiol Grx-containing TGRs. DATABASE: EC numbers: 1.8.1.9; 1.8.1.B1.


Asunto(s)
Glutatión/metabolismo , Modelos Moleculares , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Tiorredoxinas/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Biocatálisis , Dominio Catalítico , Dimerización , Disulfuros/química , Disulfuros/metabolismo , Etanol/análogos & derivados , Etanol/química , Etanol/metabolismo , Glutatión/química , Humanos , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Mutagénesis Sitio-Dirigida , Mutación , NADH NADPH Oxidorreductasas/química , NADH NADPH Oxidorreductasas/genética , Nitrobenzoatos/química , Nitrobenzoatos/metabolismo , Oxidación-Reducción , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/metabolismo , Tiorredoxinas/química
18.
Sci Rep ; 7(1): 10449, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28874682

RESUMEN

Redox balance is essential for the survival, growth and multiplication of malaria parasites and oxidative stress is involved in the mechanism of action of many antimalarial drugs. Hydrogen peroxide (H2O2) plays an important role in redox signalling and pathogen-host cell interactions. For monitoring intra- and subcellular redox events, highly sensitive and specific probes are required. Here, we stably expressed the ratiometric H2O2 redox sensor roGFP2-Orp1 in the cytosol and the mitochondria of Plasmodium falciparum (P. falciparum) NF54-attB blood-stage parasites and evaluated its sensitivity towards oxidative stress, selected antimalarial drugs, and novel lead compounds. In both compartments, the sensor showed reproducible sensitivity towards H2O2 in the low micromolar range and towards antimalarial compounds at pharmacologically relevant concentrations. Upon short-term exposure (4 h), artemisinin derivatives, quinine and mefloquine impacted H2O2 levels in mitochondria, whereas chloroquine and a glucose-6-phosphate dehydrogenase (G6PD) inhibitor affected the cytosol; 24 h exposure to arylmethylamino steroids and G6PD inhibitors revealed oxidation of mitochondria and cytosol, respectively. Genomic integration of an H2O2 sensor expressed in subcellular compartments of P. falciparum provides the basis for studying complex parasite-host cell interactions or drug effects with spatio-temporal resolution while preserving cell integrity, and sets the stage for high-throughput approaches to identify antimalarial agents perturbing redox equilibrium.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Malaria/parasitología , Oxidación-Reducción , Plasmodium/metabolismo , Antimaláricos/farmacología , Técnicas Biosensibles , Expresión Génica , Genes Reporteros , Imagen Molecular , Sondas Moleculares , Plasmodium/efectos de los fármacos , Plasmodium/genética
19.
PLoS One ; 12(4): e0174837, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28369083

RESUMEN

Hydrogen peroxide is an important antimicrobial agent but is also crucially involved in redox signaling and pathogen-host cell interactions. As a basis for systematically investigating intracellular H2O2 dynamics and regulation in living malaria parasites, we established the genetically encoded fluorescent H2O2 sensors roGFP2-Orp1 and HyPer-3 in Plasmodium falciparum. Both ratiometric redox probes as well as the pH control SypHer were expressed in the cytosol of blood-stage parasites. Both redox sensors showed reproducible sensitivity towards H2O2 in the lower micromolar range in vitro and in the parasites. Due to the pH sensitivity of HyPer-3, we used parasites expressing roGFP2-Orp1 for evaluation of short-, medium-, and long-term effects of antimalarial drugs on H2O2 levels and detoxification in Plasmodium. None of the quinolines or artemisinins tested had detectable direct effects on the H2O2 homeostasis at pharmacologically relevant concentrations. However, pre-treatment of the cells with antimalarial drugs or heat shock led to a higher tolerance towards exogenous H2O2. The systematic evaluation and comparison of the two genetically encoded cytosolic H2O2 probes in malaria parasites provides a basis for studying parasite-host cell interactions or drug effects with spatio-temporal resolution while preserving cell integrity.


Asunto(s)
Antimaláricos/farmacología , Peróxido de Hidrógeno/metabolismo , Plasmodium falciparum/efectos de los fármacos , Western Blotting , Citosol/efectos de los fármacos , Citosol/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/parasitología , Eritrocitos/fisiología , Escherichia coli , Fármacos Hematológicos/farmacología , Homeostasis/efectos de los fármacos , Calor , Humanos , Concentración de Iones de Hidrógeno , Microscopía Confocal , Oxidación-Reducción/efectos de los fármacos , Plasmodium falciparum/metabolismo , Proteínas Recombinantes/metabolismo , Saponinas/farmacología , Transfección
20.
Nat Commun ; 8: 14478, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28211535

RESUMEN

In search of antiparasitic agents, we here identify arylmethylamino steroids as potent compounds and characterize more than 60 derivatives. The lead compound 1o is fast acting and highly active against intraerythrocytic stages of chloroquine-sensitive and resistant Plasmodium falciparum parasites (IC50 1-5 nM) as well as against gametocytes. In P. berghei-infected mice, oral administration of 1o drastically reduces parasitaemia and cures the animals. Furthermore, 1o efficiently blocks parasite transmission from mice to mosquitoes. The steroid compounds show low cytotoxicity in mammalian cells and do not induce acute toxicity symptoms in mice. Moreover, 1o has a remarkable activity against the blood-feeding trematode parasite Schistosoma mansoni. The steroid and the hydroxyarylmethylamino moieties are essential for antimalarial activity supporting a chelate-based quinone methide mechanism involving metal or haem bioactivation. This study identifies chemical scaffolds that are rapidly internalized into blood-feeding parasites.


Asunto(s)
Aminas/farmacología , Antiparasitarios/farmacología , Esteroides/farmacología , Aminas/química , Aminas/farmacocinética , Animales , Anopheles/parasitología , Antiinfecciosos/farmacología , Antiparasitarios/química , Antiparasitarios/farmacocinética , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Células Germinativas/efectos de los fármacos , Concentración 50 Inhibidora , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria/parasitología , Malaria/transmisión , Ratones , Modelos Biológicos , Parásitos/efectos de los fármacos , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/crecimiento & desarrollo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/ultraestructura , Esteroides/química , Esteroides/farmacocinética , Pruebas de Toxicidad Aguda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA