Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 285(27): 20827-33, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20439460

ABSTRACT

The survival and proliferation of the obligate intracellular malaria parasite Plasmodium falciparum require salvage of essential purines from the host. Genetic studies have previously shown that the parasite plasma membrane purine permease, PfNT1, plays an essential function in the transport of all naturally occurring purine nucleosides and nucleobases across the parasite plasma membrane. Here, we describe an intracellular permease, PfNT2. PfNT2 is, like PfNT1, a member of the equilibrative nucleoside transporter family. Confocal and immunoelectron microscopic analyses of transgenic parasites harboring green fluorescent protein- or hemagglutinin-tagged PfNT2 demonstrated endoplasmic reticulum localization. This localization was confirmed by colocalization with the endoplasmic reticulum marker PfBiP. Using yeast as a surrogate system, we show that targeting PfNT2 to the plasma membrane of fui1Delta cells lacking the plasma membrane nucleoside transporter Fui1 confers sensitivity to the toxic nucleoside analog 5-fluorouridine. This study provides the first evidence of an intracellular purine permease in apicomplexan parasites and suggests a novel biological function for the parasite endoplasmic reticulum during malaria infection.


Subject(s)
Endoplasmic Reticulum/enzymology , Membrane Transport Proteins/genetics , Nucleoside Transport Proteins/genetics , Plasmodium falciparum/enzymology , Amino Acid Sequence , Animals , Endoplasmic Reticulum/ultrastructure , Erythrocytes/parasitology , Floxuridine/metabolism , Genes, Reporter , Host-Parasite Interactions , Humans , Malaria, Falciparum/blood , Membrane Transport Proteins/metabolism , Microscopy, Immunoelectron , Nucleoside Transport Proteins/metabolism , Parasitemia/blood , Plasmodium falciparum/genetics , Promoter Regions, Genetic , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Purines/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Transfection
2.
Cell Microbiol ; 12(7): 930-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20088947

ABSTRACT

Repeated immunizations with whole Plasmodium blood stage parasites and concomitant drug cure of infection confer protective immunity against parasite challenge in mice, monkeys and humans. Moreover, it was recently shown that infections with genetically modified rodent malaria blood stage parasites conferred sterile protection against lethal blood stage challenge. However, in these models vaccination resulted in high parasitemias and, in consequence, carries risk of vaccine-induced pathology and death. Herein, we generated a novel, completely blood stage-attenuated P. yoelii rodent malaria strain by targeted deletion of parasite nucleoside transporter 1 (NT1). Immunization of inbred and outbred mouse strains with a single low dose of Pynt1(-) blood stages did not induce any patent infections and conferred complete sterile protection against lethal heterologous blood stage and sporozoite challenges. Partial protection was observed against lethal challenges with another parasite species, P. berghei. Importantly, subcutaneous immunization with Pynt1(-) conferred sterile protection against lethal blood stage challenges. We show that cellular and humoral immune responses are both essential for sterile protection. The study demonstrates that genetic manipulation provides a platform for the designed, complete attenuation of malaria parasite blood stages and suggests testing the safety and efficacy of P. falciparum NT1 knockout strains in humans.


Subject(s)
Malaria/immunology , Malaria/parasitology , Nucleoside Transport Proteins/genetics , Plasmodium/immunology , Protozoan Proteins/genetics , Animals , Female , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium/genetics , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology
3.
Trends Parasitol ; 25(6): 246-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19423394

ABSTRACT

In a recent paper, Quashie et al. have proposed that purine uptake into the intraerythrocytic malaria parasite involves four different plasma membrane transporters - two high affinity and two low affinity. They equate one of the two high-affinity transporters with PfNT1, a transporter reported previously to be a low-affinity system. Here, we offer an alternative interpretation of their data, suggesting that the conclusions drawn by Quashie et al. take insufficient account of metabolism.


Subject(s)
Membrane Transport Proteins/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Purines/metabolism , Animals , Biological Transport , Oocytes/metabolism , Xenopus laevis/parasitology
4.
Mol Biochem Parasitol ; 161(2): 130-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18639591

ABSTRACT

The malaria parasite, Plasmodium falciparum, is unable to synthesize the purine ring de novo and is therefore wholly dependent upon purine salvage from the host for survival. Previous studies have indicated that a P. falciparum strain in which the purine transporter PfNT1 had been disrupted was unable to grow on physiological concentrations of adenosine, inosine and hypoxanthine. We have now used an episomally complemented pfnt1Delta knockout parasite strain to confirm genetically the functional role of PfNT1 in P. falciparum purine uptake and utilization. Episomal complementation by PfNT1 restored the ability of pfnt1Delta parasites to transport and utilize adenosine, inosine and hypoxanthine as purine sources. The ability of wild-type and pfnt1Delta knockout parasites to transport and utilize the other physiologically relevant purines adenine, guanine, guanosine and xanthine was also examined. Unlike wild-type and complemented P. falciparum parasites, pfnt1Delta parasites could not proliferate on guanine, guanosine or xanthine as purine sources, and no significant transport of these substrates could be detected in isolated parasites. Interestingly, whereas isolated pfnt1Delta parasites were still capable of adenine transport, these parasites grew only when adenine was provided at high, non-physiological concentrations. Taken together these results demonstrate that, in addition to hypoxanthine, inosine and adenosine, PfNT1 is essential for the transport and utilization of xanthine, guanine and guanosine.


Subject(s)
Biological Transport , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Purines/metabolism , Adenine/metabolism , Animals , Erythrocytes/parasitology , Gene Deletion , Guanine/metabolism , Guanosine/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Purines/chemistry , Xanthine/metabolism
6.
Int J Parasitol ; 38(2): 203-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17765902

ABSTRACT

Hypoxanthine, a nucleobase, serves as the major source of the essential purine group for the intraerythrocytic malaria parasite. In this study we have measured the uptake of hypoxanthine, and that of the related purine nucleobase adenine, by mature blood-stage Plasmodium falciparum parasites isolated from their host cells by saponin-permeabilisation of the erythrocyte and parasitophorous vacuole membranes. The uptake of both [3H]hypoxanthine and [3H]adenine was comprised of at least two components; in each case there was a rapid equilibration of the radiolabel between the intra- and extracellular solutions via a low-affinity transport mechanism, and an accumulation of radiolabel (such that the estimated intracellular concentration exceeded the extracellular concentration) via a higher-affinity process. The uptake of [3H]adenine was studied in more detail. The rapid, low-affinity equilibration of [3H]adenine between the intra-and extracellular solution was independent of the energy status of the parasite whereas the higher-affinity accumulation of the radiolabel was ATP-dependent. A kinetic analysis of adenine uptake revealed that the low-affinity (equilibrative) process had a Km of approximately 1.2mM, similar to the value of 0.82 mM estimated here (using the Xenopus laevis oocyte expression system) for the Km for the transport of adenine by PfENT1, a parasite-encoded member of the 'equilibrative nucleoside/nucleobase transporter' family. The results indicate that nucleobases enter the intraerythrocytic parasite via a rapid, equilibrative process that has kinetic characteristics similar to those of PfENT1.


Subject(s)
Erythrocytes/parasitology , Nucleobase Transport Proteins/metabolism , Plasmodium falciparum/metabolism , Adenine/analysis , Adenine/metabolism , Adenosine Triphosphate/analysis , Adenosine Triphosphate/metabolism , Animals , Biological Transport , Cells, Cultured , Hypoxanthine/analysis , Hypoxanthine/metabolism , Malaria, Falciparum , Nucleobase Transport Proteins/analysis , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/metabolism , Oocytes/metabolism , Parasitology/methods , Protozoan Proteins/metabolism , Xenopus laevis
7.
Nature ; 443(7111): 582-5, 2006 Oct 05.
Article in English | MEDLINE | ID: mdl-17006451

ABSTRACT

As the malaria parasite, Plasmodium falciparum, grows within its host erythrocyte it induces an increase in the permeability of the erythrocyte membrane to a range of low-molecular-mass solutes, including Na+ and K+ (ref. 1). This results in a progressive increase in the concentration of Na+ in the erythrocyte cytosol. The parasite cytosol has a relatively low Na+ concentration and there is therefore a large inward Na+ gradient across the parasite plasma membrane. Here we show that the parasite exploits the Na+ electrochemical gradient to energize the uptake of inorganic phosphate (P(i)), an essential nutrient. P(i) was taken up into the intracellular parasite by a Na+-dependent transporter, with a stoichiometry of 2Na+:1P(i) and with an apparent preference for the monovalent over the divalent form of P(i). A P(i) transporter (PfPiT) belonging to the PiT family was cloned from the parasite and localized to the parasite surface. Expression of PfPiT in Xenopus oocytes resulted in Na+-dependent P(i) uptake with characteristics similar to those observed for P(i) uptake in the parasite. This study provides new insight into the significance of the malaria-parasite-induced alteration of the ionic composition of its host cell.


Subject(s)
Malaria/parasitology , Phosphate Transport Proteins/metabolism , Phosphates/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Sodium/pharmacology , Animals , Biological Transport/drug effects , Erythrocytes/drug effects , Erythrocytes/parasitology , Hydrogen-Ion Concentration , Kinetics , Oocytes , Phylogeny , Saponins/pharmacology , Xenopus
8.
Mol Microbiol ; 60(3): 738-48, 2006 May.
Article in English | MEDLINE | ID: mdl-16629674

ABSTRACT

Like all parasitic protozoa, the human malaria parasite Plasmodium falciparum lacks the enzymes required for de novo synthesis of purines and it is therefore reliant upon the salvage of these compounds from the external environment. P. falciparum equilibrative nucleoside transporter 1 (PfENT1) is a nucleoside transporter that has been localized to the plasma membrane of the intraerythrocytic form of the parasite. In this study we have characterized the transport of purine and pyrimidine nucleosides across the plasma membrane of 'isolated' trophozoite-stage P. falciparum parasites and compared the transport characteristics of the parasite with those of PfENT1 expressed in Xenopus oocytes. The transport of nucleosides into the parasite: (i) was, in the case of adenosine, inosine and thymidine, very fast, equilibrating within a few seconds; (ii) was of low affinity [K(m) (adenosine) = 1.45 +/- 0.25 mM; K(m) (thymidine) = 1.11 +/- 0.09 mM]; and (iii) showed 'cross-competition' for adenosine, inosine and thymidine, but not cytidine. The kinetic characteristics of nucleoside transport in intact parasites matched very closely those of PfENT1 expressed in Xenopus oocytes [K(m) (adenosine) = 1.86 +/- 0.28 mM; K(m) (thymidine) = 1.33 +/- 0.17 mM]. Furthermore, PfENT1 transported adenosine, inosine and thymidine, with a cross-competition profile the same as that seen for isolated parasites. The data are consistent with PfENT1 serving as a major route for the uptake of nucleosides across the parasite plasma membrane.


Subject(s)
Cell Membrane/metabolism , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Purines/metabolism , Pyrimidines/metabolism , Animals , Erythrocytes/parasitology , Humans , Nucleoside Transport Proteins/metabolism , Oocytes/metabolism , Plasmodium falciparum/growth & development , Xenopus/growth & development , Xenopus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...