Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(30): e2306420120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37463201

ABSTRACT

To ensure their survival in the human bloodstream, malaria parasites degrade up to 80% of the host erythrocyte hemoglobin in an acidified digestive vacuole. Here, we combine conditional reverse genetics and quantitative imaging approaches to demonstrate that the human malaria pathogen Plasmodium falciparum employs a heteromultimeric V-ATPase complex to acidify the digestive vacuole matrix, which is essential for intravacuolar hemoglobin release, heme detoxification, and parasite survival. We reveal an additional function of the membrane-embedded V-ATPase subunits in regulating morphogenesis of the digestive vacuole independent of proton translocation. We further show that intravacuolar accumulation of antimalarial chemotherapeutics is surprisingly resilient to severe deacidification of the vacuole and that modulation of V-ATPase activity does not affect parasite sensitivity toward these drugs.


Subject(s)
Antimalarials , Malaria, Falciparum , Parasites , Animals , Humans , Antimalarials/pharmacology , Antimalarials/metabolism , Adenosine Triphosphatases/metabolism , Vacuoles , Malaria, Falciparum/parasitology , Plasmodium falciparum/metabolism
2.
Nat Commun ; 14(1): 4234, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37454114

ABSTRACT

The chloroquine resistance transporter, PfCRT, of the human malaria parasite Plasmodium falciparum is sensitive to acidic pH. Consequently, PfCRT operates at 60% of its maximal drug transport activity at the pH of 5.2 of the digestive vacuole, a proteolytic organelle from which PfCRT expels drugs interfering with heme detoxification. Here we show by alanine-scanning mutagenesis that E207 is critical for pH sensing. The E207A mutation abrogates pH-sensitivity, while preserving drug substrate specificity. Substituting E207 with Asp or His, but not other amino acids, restores pH-sensitivity. Molecular dynamics simulations and kinetics analyses suggest an allosteric binding model in which PfCRT can accept both protons and chloroquine in a partial noncompetitive manner, with increased proton concentrations decreasing drug transport. Further simulations reveal that E207 relocates from a peripheral to an engaged location during the transport cycle, forming a salt bridge with residue K80. We propose that the ionized carboxyl group of E207 acts as a hydrogen acceptor, facilitating transport cycle progression, with pH sensing as a by-product.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Antimalarials/pharmacology , Antimalarials/chemistry , Chloroquine/pharmacology , Membrane Transport Proteins/metabolism , Protozoan Proteins/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Hydrogen-Ion Concentration , Drug Resistance/genetics , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology
3.
PLoS Pathog ; 19(6): e1011436, 2023 06.
Article in English | MEDLINE | ID: mdl-37285379

ABSTRACT

The chloroquine resistance transporter (PfCRT) confers resistance to a wide range of quinoline and quinoline-like antimalarial drugs in Plasmodium falciparum, with local drug histories driving its evolution and, hence, the drug transport specificities. For example, the change in prescription practice from chloroquine (CQ) to piperaquine (PPQ) in Southeast Asia has resulted in PfCRT variants that carry an additional mutation, leading to PPQ resistance and, concomitantly, to CQ re-sensitization. How this additional amino acid substitution guides such opposing changes in drug susceptibility is largely unclear. Here, we show by detailed kinetic analyses that both the CQ- and the PPQ-resistance conferring PfCRT variants can bind and transport both drugs. Surprisingly, the kinetic profiles revealed subtle yet significant differences, defining a threshold for in vivo CQ and PPQ resistance. Competition kinetics, together with docking and molecular dynamics simulations, show that the PfCRT variant from the Southeast Asian P. falciparum strain Dd2 can accept simultaneously both CQ and PPQ at distinct but allosterically interacting sites. Furthermore, combining existing mutations associated with PPQ resistance created a PfCRT isoform with unprecedented non-Michaelis-Menten kinetics and superior transport efficiency for both CQ and PPQ. Our study provides additional insights into the organization of the substrate binding cavity of PfCRT and, in addition, reveals perspectives for PfCRT variants with equal transport efficiencies for both PPQ and CQ.


Subject(s)
Antimalarials , Malaria, Falciparum , Plasmodium falciparum , Quinolines , Humans , Antimalarials/chemistry , Chloroquine/pharmacology , Chloroquine/therapeutic use , Drug Resistance/genetics , Kinetics , Malaria, Falciparum/drug therapy , Mutation , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use
4.
Redox Biol ; 58: 102536, 2022 12.
Article in English | MEDLINE | ID: mdl-36401887

ABSTRACT

The redox state of the host-parasite unit has been hypothesized to play a central role for the fitness of the intraerythrocytic blood stages of the human malaria parasite Plasmodium falciparum. In particular, hemoglobinopathies have been suggested to cause a more oxidizing environment, thereby protecting from severe malaria. Here we determined the redox potential of infected wild-type (hemoglobin AA) or sickle trait (hemoglobin AS) erythrocytes using parasite-encoded variants of the redox-sensitive green-fluorescent protein 2 (roGFP2). Our non-invasive roGFP2 single-cell measurements revealed a reducing steady-state redox potential of -304 ± 11 mV for the erythrocyte cytosol during ring-stage development and a rather sudden oxidation to -278 ± 12 mV during trophozoite-stage development around 28 h post invasion. There was no significant difference between wild-type or sickle trait erythrocytes regarding the stage dependence and the detected increase of the redox potential during the intraerythrocytic life cycle. The steady-state redox potential of the parasite cytosol, between -304 and -313 mV, was highly reducing throughout the life cycle. The redox potential in the parasitophorous vacuole at the interface between the secretory pathway and the erythrocyte was -284 ± 10 mV and remained stable during trophozoite-stage development with implications for the export of disulfide-containing proteins. In summary, P. falciparum blood stage development from the late ring to the early trophozoite stage causes a physiological jump in erythrocyte redox potential irrespective of the presence or absence of hemoglobin S.


Subject(s)
Malaria, Falciparum , Sickle Cell Trait , Humans , Plasmodium falciparum/metabolism , Sickle Cell Trait/genetics , Sickle Cell Trait/metabolism , Erythrocytes/metabolism , Malaria, Falciparum/parasitology , Green Fluorescent Proteins/metabolism , Oxidation-Reduction
5.
Microbiol Spectr ; 10(4): e0110122, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35867395

ABSTRACT

The chloroquine resistance transporter, PfCRT, is an essential factor during intraerythrocytic development of the human malaria parasite Plasmodium falciparum. PfCRT resides at the digestive vacuole of the parasite, where hemoglobin taken up by the parasite from its host cell is degraded. PfCRT can acquire several mutations that render PfCRT a drug transporting system expelling compounds targeting hemoglobin degradation from the digestive vacuole. The non-drug related function of PfCRT is less clear, although a recent study has suggested a role in oligopeptide transport based on studies conducted in a heterologous expression system. The uncertainty about the natural function of PfCRT is partly due to a lack of a null mutant and a dearth of functional assays in the parasite. Here, we report on the generation of a conditional PfCRT knock-down mutant in P. falciparum. The mutant accumulated oligopeptides 2 to at least 8 residues in length under knock-down conditions, as shown by comparative global metabolomics. The accumulated oligopeptides were structurally diverse, had an isoelectric point between 4.0 and 5.4 and were electrically neutral or carried a single charge at the digestive vacuolar pH of 5.2. Fluorescently labeled dipeptides and live cell imaging identified the digestive vacuole as the compartment where oligopeptides accumulated. Our findings suggest a function of PfCRT in oligopeptide transport across the digestive vacuolar membrane in P. falciparum and associated with it a role in nutrient acquisition and the maintenance of the colloid osmotic balance. IMPORTANCE The chloroquine resistance transporter, PfCRT, is important for the survival of the human malaria parasite Plasmodium falciparum. It increases the tolerance to many antimalarial drugs, and it is essential for the development of the parasite within red blood cells. While we understand the role of PfCRT in drug resistance in ever increasing detail, the non-drug resistance functions are still debated. Identifying the natural substrate of PfCRT has been hampered by a paucity of functional assays to test putative substrates in the parasite system and the absence of a parasite mutant deficient for the PfCRT encoding gene. By generating a conditional PfCRT knock-down mutant, together with comparative metabolomics and uptake studies using fluorescently labeled oligopeptides, we could show that PfCRT is an oligopeptide transporter. The oligopeptides were structurally diverse and were electrically neutral or carried a single charge. Our data support a function of PfCRT in oligopeptide transport.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria , Antimalarials/pharmacology , Chloroquine/metabolism , Chloroquine/pharmacology , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Oligopeptides/metabolism , Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
6.
Methods Mol Biol ; 2470: 457-466, 2022.
Article in English | MEDLINE | ID: mdl-35881366

ABSTRACT

Super-resolution microscopy in the form of photoactivated localization microscopy (PALM) offers the possibility of counting single molecules in a cell, a cellular compartment or a molecular complex. PALM can, therefore, underpin molecular and biochemical processes with a numeric and stoichiometric understanding of the interacting players. Here, we introduce the physical principles underlying PALM and provide a step-by-step protocol of how to apply PALM to questions related to the biology and pathophysiology of P. falciparum and other malaria parasites.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Antigen Presentation , Erythrocytes/parasitology , Humans , Malaria, Falciparum/parasitology , Microscopy , Protozoan Proteins/chemistry
7.
PLoS Comput Biol ; 18(4): e1009509, 2022 04.
Article in English | MEDLINE | ID: mdl-35394995

ABSTRACT

Red blood cells can withstand the harsh mechanical conditions in the vasculature only because the bending rigidity of their plasma membrane is complemented by the shear elasticity of the underlying spectrin-actin network. During an infection by the malaria parasite Plasmodium falciparum, the parasite mines host actin from the junctional complexes and establishes a system of adhesive knobs, whose main structural component is the knob-associated histidine rich protein (KAHRP) secreted by the parasite. Here we aim at a mechanistic understanding of this dramatic transformation process. We have developed a particle-based computational model for the cytoskeleton of red blood cells and simulated it with Brownian dynamics to predict the mechanical changes resulting from actin mining and KAHRP-clustering. Our simulations include the three-dimensional conformations of the semi-flexible spectrin chains, the capping of the actin protofilaments and several established binding sites for KAHRP. For the healthy red blood cell, we find that incorporation of actin protofilaments leads to two regimes in the shear response. Actin mining decreases the shear modulus, but knob formation increases it. We show that dynamical changes in KAHRP binding affinities can explain the experimentally observed relocalization of KAHRP from ankyrin to actin complexes and demonstrate good qualitative agreement with experiments by measuring pair cross-correlations both in the computer simulations and in super-resolution imaging experiments.


Subject(s)
Malaria , Protozoan Proteins , Actins/metabolism , Cytoskeleton/metabolism , Erythrocyte Membrane , Erythrocytes/metabolism , Humans , Peptides/chemistry , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Spectrin
8.
Mol Microbiol ; 117(2): 274-292, 2022 02.
Article in English | MEDLINE | ID: mdl-34514656

ABSTRACT

The knob-associated histidine-rich protein (KAHRP) plays a pivotal role in the pathophysiology of Plasmodium falciparum malaria by forming membrane protrusions in infected erythrocytes, which anchor parasite-encoded adhesins to the membrane skeleton. The resulting sequestration of parasitized erythrocytes in the microvasculature leads to severe disease. Despite KAHRP being an important virulence factor, its physical location within the membrane skeleton is still debated, as is its function in knob formation. Here, we show by super-resolution microscopy that KAHRP initially associates with various skeletal components, including ankyrin bridges, but eventually colocalizes with remnant actin junctions. We further present a 35 Å map of the spiral scaffold underlying knobs and show that a KAHRP-targeting nanoprobe binds close to the spiral scaffold. Single-molecule localization microscopy detected ~60 KAHRP molecules/knob. We propose a dynamic model of KAHRP organization and a function of KAHRP in attaching other factors to the spiral scaffold.


Subject(s)
Actins , Plasmodium falciparum , Actins/metabolism , Erythrocytes/metabolism , Histidine/metabolism , Peptides/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism
9.
PLoS Pathog ; 17(10): e1009969, 2021 10.
Article in English | MEDLINE | ID: mdl-34614006

ABSTRACT

The pathology associated with malaria infection is largely due to the ability of infected human RBCs to adhere to a number of receptors on endothelial cells within tissues and organs. This phenomenon is driven by the export of parasite-encoded proteins to the host cell, the exact function of many of which is still unknown. Here we inactivate the function of one of these exported proteins, PFA66, a member of the J-domain protein family. Although parasites lacking this protein were still able to grow in cell culture, we observed severe defects in normal host cell modification, including aberrant morphology of surface knobs, disrupted presentation of the cytoadherence molecule PfEMP1, and a total lack of cytoadherence, despite the presence of the knob associated protein KAHRP. Complementation assays demonstrate that an intact J-domain is required for recovery to a wild-type phenotype and suggest that PFA66 functions in concert with a HSP70 to carry out host cell modification. Strikingly, this HSP70 is likely to be of host origin. ATPase assays on recombinant protein verify a functional interaction between PFA66 and residual host cell HSP70. Taken together, our data reveal a role for PFA66 in host cell modification, strongly implicate human HSP70s as being essential in this process and uncover a new KAHRP-independent molecular factor required for correct knob biogenesis.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Host-Parasite Interactions/physiology , Malaria, Falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Plasmodium falciparum/metabolism , Virulence
10.
Biophys J ; 120(16): 3315-3328, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34246628

ABSTRACT

The pathology of Plasmodium falciparum malaria is largely defined by the cytoadhesion of infected erythrocytes to the microvascular endothelial lining. The complexity of the endothelial surface and the large range of interactions available for the infected erythrocyte via parasite-encoded adhesins make analysis of critical contributions during cytoadherence challenging to define. Here, we have explored supported membranes functionalized with two important adhesion receptors, ICAM1 or CD36, as a quantitative biomimetic surface to help understand the processes involved in cytoadherence. Parasitized erythrocytes bound to the receptor-functionalized membranes with high efficiency and selectivity under both static and flow conditions, with infected wild-type erythrocytes displaying a higher binding capacity than do parasitized heterozygous sickle cells. We further show that the binding efficiency decreased with increasing intermolecular receptor distance and that the cell-surface contacts were highly dynamic and increased with rising wall shear stress as the cell underwent a shape transition. Computer simulations using a deformable cell model explained the wall-shear-stress-induced dynamic changes in cell shape and contact area via the specific physical properties of erythrocytes, the density of adhesins presenting knobs, and the lateral movement of receptors in the supported membrane.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , CD36 Antigens , Cell Adhesion , Erythrocytes/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism
11.
Sci Rep ; 9(1): 18333, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31797898

ABSTRACT

The glms ribozyme system has been used as an amenable tool to conditionally control expression of genes of interest. It is generally assumed that insertion of the ribozyme sequence does not affect expression of the targeted gene in the absence of the inducer glucosamine-6-phosphate, although experimental support for this assumption is scarce. Here, we report the unexpected finding that integration of the glms ribozyme sequence in the 3' untranslated region of a gene encoding a HECT E3 ubiquitin ligase, termed Plasmodium falciparum ubiquitin transferase (PfUT), increased steady state RNA and protein levels 2.5-fold in the human malaria parasite P. falciparum. Overexpression of pfut resulted in an S/M phase-associated lengthening of the parasite's intraerythrocytic developmental cycle and a reduced merozoite invasion efficiency. The addition of glucosamine partially restored the wild type phenotype. Our study suggests a role of PfUT in controlling cell cycle progression and merozoite invasion. Our study further raises awareness regarding unexpected effects on gene expression when inserting the glms ribozyme sequence into a gene locus.


Subject(s)
Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin/genetics , Erythrocytes/enzymology , Gene Expression Regulation, Enzymologic/genetics , Glucosamine/analogs & derivatives , Glucosamine/metabolism , Glucose-6-Phosphate/analogs & derivatives , Glucose-6-Phosphate/metabolism , Humans , Malaria, Falciparum/pathology , Phenotype , Plasmodium falciparum/enzymology , Plasmodium falciparum/pathogenicity
12.
Commun Biol ; 2: 311, 2019.
Article in English | MEDLINE | ID: mdl-31428699

ABSTRACT

During intraerythrocytic development, the human malaria parasite Plasmodium falciparum alters the mechanical deformability of its host cell. The underpinning biological processes involve gain in parasite mass, changes in the membrane protein compositions, reorganization of the cytoskeletons and its coupling to the plasma membrane, and formation of membrane protrusions, termed knobs. The hemoglobinopathies S and C are known to partially protect carriers from severe malaria, possibly through additional changes in the erythrocyte biomechanics, but a detailed quantification of cell mechanics is still missing. Here, we combined flicker spectroscopy and a mathematical model and demonstrated that knob formation strongly suppresses membrane fluctuations by increasing membrane-cytoskeleton coupling. We found that the confinement increased with hemoglobin S but decreases with hemoglobin C in spite of comparable knob densities and diameters. We further found that the membrane bending modulus strongly depends on the hemoglobinopathetic variant, suggesting increased amounts of irreversibly oxidized hemichromes bound to membranes.


Subject(s)
Erythrocyte Membrane/parasitology , Hemoglobin C/metabolism , Hemoglobin, Sickle/metabolism , Plasmodium falciparum/physiology , Biomechanical Phenomena , Computer Simulation , Humans , Mutation/genetics , Numerical Analysis, Computer-Assisted
13.
J Biol Chem ; 294(34): 12766-12778, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31285265

ABSTRACT

The chloroquine resistance transporter PfCRT of the human malaria parasite Plasmodium falciparum confers resistance to the former first-line antimalarial drug chloroquine, and it modulates the responsiveness to a wide range of quinoline and quinoline-like compounds. PfCRT is post-translationally modified by phosphorylation, palmitoylation, and, possibly, ubiquitination. However, the impact of these post-translational modifications on P. falciparum biology and, in particular, the drug resistance-conferring activity of PfCRT has remained elusive. Here, we confirm phosphorylation at Ser-33 and Ser-411 of PfCRT of the chloroquine-resistant P. falciparum strain Dd2 and show that kinase inhibitors can sensitize drug responsiveness. Using CRISPR/Cas9 genome editing to generate genetically engineered PfCRT variants in the parasite, we further show that substituting Ser-33 with alanine reduced chloroquine and quinine resistance by ∼50% compared with the parental P. falciparum strain Dd2, whereas the phosphomimetic amino acid aspartic acid could fully and glutamic acid could partially reconstitute the level of chloroquine/quinine resistance. Transport studies conducted in the parasite and in PfCRT-expressing Xenopus laevis oocytes linked phosphomimetic substitution at Ser-33 to increased transport velocity. Our data are consistent with phosphorylation of Ser-33 relieving an autoinhibitory intramolecular interaction within PfCRT, leading to a stimulated drug transport activity. Our findings shed additional light on the function of PfCRT and suggest that chloroquine could be reevaluated as an antimalarial drug by targeting the kinase in P. falciparum that phosphorylates Ser-33 of PfCRT.


Subject(s)
Membrane Transport Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Serine/metabolism , Antimalarials/pharmacology , Chloroquine/pharmacology , Drug Resistance/drug effects , Kinetics , Parasitic Sensitivity Tests , Phosphorylation , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors
14.
Commun Biol ; 2: 172, 2019.
Article in English | MEDLINE | ID: mdl-31098405

ABSTRACT

PfEMP1 (erythrocyte membrane protein 1) adhesins play a pivotal role in the pathophysiology of falciparum malaria, by mediating sequestration of Plasmodium falciparum-infected erythrocytes in the microvasculature. PfEMP1 variants are expressed by var genes and are presented on membrane elevations, termed knobs. However, the organization of PfEMP1 on knobs is largely unclear. Here, we use super-resolution microscopy and genetically altered parasites expressing a modified var2csa gene in which the coding sequence of the photoactivatable mEOS2 was inserted to determine the number and distribution of PfEMP1 on single knobs. The data were verified by quantitative fluorescence-activated cell sorting analysis and immuno-electron microscopy together with stereology methods. We show that knobs contain 3.3 ± 1.7 and 4.3 ± 2.5 PfEMP1 molecules, predominantly placed on the knob tip, in parasitized erythrocytes containing wild type and sickle haemoglobin, respectively. The ramifications of our findings for cytoadhesion and immune evasion are discussed.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Erythrocytes/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Animals , Antigens, Protozoan/chemistry , Erythrocytes/immunology , Erythrocytes/ultrastructure , Genes, Protozoan , Genetic Variation , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Microscopy, Immunoelectron , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single Molecule Imaging
15.
Commun Biol ; 1: 211, 2018.
Article in English | MEDLINE | ID: mdl-30534603

ABSTRACT

Sickle cell trait, a common hereditary blood disorder, protects carriers from severe disease in infections with the human malaria parasite Plasmodium falciparum. Protection is associated with a reduced capacity of parasitized erythrocytes to cytoadhere to the microvascular endothelium and cause vaso-occlusive events. However, the underpinning cellular and biomechanical processes are only partly understood and the impact on endothelial cell activation is unclear. Here, we show, by combining quantitative flow chamber experiments with multiscale computer simulations of deformable cells in hydrodynamic flow, that parasitized erythrocytes containing the sickle cell haemoglobin displayed altered adhesion dynamics, resulting in restricted contact footprints on the endothelium. Main determinants were cell shape, knob density and membrane bending. As a consequence, the extent of endothelial cell activation was decreased. Our findings provide a quantitative understanding of how the sickle cell trait affects the dynamic cytoadhesion behavior of parasitized erythrocytes and, in turn, endothelial cell activation.

16.
Malar J ; 17(1): 121, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29558913

ABSTRACT

BACKGROUND: The increased resistance of the human malaria parasite Plasmodium falciparum to currently employed drugs creates an urgent call for novel anti-malarial drugs. Particularly, efforts should be devoted to developing fast-acting anti-malarial compounds in case clinical resistance increases to the first-line artemisinin-based combination therapy. SC83288, an amicarbalide derivative, is a clinical development candidate for the treatment of severe malaria. SC83288 is fast-acting and able to clear P. falciparum parasites at low nanomolar concentrations in vitro, as well as in a humanized SCID mouse model system in vivo. In this study, the antiplasmodial activity of SC83288 against artemisinins was profiled in order to assess its potential to replace, or be combined with, artemisinin derivatives. RESULTS: Based on growth inhibition and ring survival assays, no cross-resistance was observed between artemisinins and SC83288, using parasite lines that were resistant to either one of these drugs. In addition, no synergistic or antagonistic interaction was observed between the two drugs. This study further confirmed that SC83288 is a fast acting drug in several independent assays. Combinations of SC83288 and artesunate maintained the rapid parasite killing activities of both components. CONCLUSION: The results obtained in this study are consistent with artemisinins and SC83288 having distinct modes of action and different mechanisms of resistance. This study further supports efforts to continue the clinical development of SC83288 against severe malaria as an alternative to artemisinins in areas critically affected by artemisinin-resistance. Considering its fast antiplasmodial activity, SC83288 could be combined with a slow-acting anti-malarial drug.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Carbanilides/pharmacology , Plasmodium falciparum/drug effects , Animals , Antimalarials/administration & dosage , Antimalarials/pharmacokinetics , Artemisinins/administration & dosage , Artemisinins/pharmacokinetics , Carbanilides/administration & dosage , Carbanilides/pharmacokinetics , Drug Interactions , Drug Resistance , Molecular Structure
17.
J Biol Chem ; 292(39): 16109-16121, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28768767

ABSTRACT

The chloroquine resistance transporter of the human malaria parasite Plasmodium falciparum, PfCRT, is an important determinant of resistance to several quinoline and quinoline-like antimalarial drugs. PfCRT also plays an essential role in the physiology of the parasite during development inside erythrocytes. However, the function of this transporter besides its role in drug resistance is still unclear. Using electrophysiological and flux experiments conducted on PfCRT-expressing Xenopus laevis oocytes, we show here that both wild-type PfCRT and a PfCRT variant associated with chloroquine resistance transport both ferrous and ferric iron, albeit with different kinetics. In particular, we found that the ability to transport ferrous iron is reduced by the specific polymorphisms acquired by the PfCRT variant as a result of chloroquine selection. We further show that iron and chloroquine transport via PfCRT is electrogenic. If these findings in the Xenopus model extend to P. falciparum in vivo, our data suggest that PfCRT might play a role in iron homeostasis, which is essential for the parasite's development in erythrocytes.


Subject(s)
Antimalarials/metabolism , Chloroquine/metabolism , Iron/metabolism , Membrane Transport Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Amino Acid Substitution , Animals , Biological Transport , Iron/chemistry , Kinetics , Membrane Transport Proteins/genetics , Mutation , Oocytes/metabolism , Oxidation-Reduction , Patch-Clamp Techniques , Protozoan Proteins/genetics , Recombinant Proteins/metabolism , Xenopus laevis
18.
Sci Rep ; 7(1): 4410, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28667301

ABSTRACT

Artemisinins are the current mainstay of malaria chemotherapy. Their exact mode of action is an ongoing matter of debate, and several factors have recently been reported to affect an early stage of artemisinin resistance of the most important human malaria parasite Plasmodium falciparum. Here, we identified a locus on chromosome 7 that affects the artemisinin susceptibility of P. falciparum in a quantitative trait locus analysis of a genetic cross between strains 7G8 and GB4. This locus includes the peroxiredoxin gene PFAOP. However, steady-state kinetic data with recombinant PfAOP do not support a direct interaction between this peroxidase and the endoperoxide artemisinin. Furthermore, neither the overexpression nor the deletion of the encoding gene affected the IC50 values for artemisinin or the oxidants diamide and tert-butyl hydroperoxide. Thus, PfAOP is dispensable for blood stage parasite survival, and the correlation between the artemisinin susceptibility and chromosome 7 is probably based on another gene within the identified locus.


Subject(s)
Artemisinins/pharmacology , Peroxiredoxins/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Artemisinins/chemistry , Chromosome Mapping , Gene Expression , Gene Knockout Techniques , Humans , Malaria, Falciparum/parasitology , Parasitic Sensitivity Tests , Peroxiredoxins/antagonists & inhibitors , Quantitative Trait Loci
20.
ACS Infect Dis ; 3(2): 119-131, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183182

ABSTRACT

Mutational changes in the Plasmodium falciparum chloroquine resistance transporter (PfCRT) have been associated with differential responses to a wide spectrum of biologically active compounds including current and former quinoline and quinoline-like antimalarial drugs. PfCRT confers altered drug responsiveness by acting as a transport system, expelling drugs from the parasite's digestive vacuole where these drugs exert, at least part of, their antiplasmodial activity. To preserve the efficacy of these invaluable drugs, novel functional tools are required for epidemiological surveys of parasite strains carrying mutant PfCRT variants and for drug development programs aimed at inhibiting or circumventing the action of PfCRT. Here we report the synthesis and characterization of a pH-sensitive fluorescent chloroquine analogue consisting of 7-chloro-N-{2-[(propan-2-yl)amino]ethyl}quinolin-4-amine functionalized with the fluorochrome 7-nitrobenzofurazan (NBD) (henceforth termed Fluo-CQ). In the parasite, Fluo-CQ accumulates in the digestive vacuole, giving rise to a strong fluorescence signal but only in parasites carrying the wild type PfCRT. In parasites carrying the mutant PfCRT, Fluo-CQ does not accumulate. The differential handling of the fluorescent probe, combined with live cell imaging, provides a diagnostic tool for quick detection of those P. falciparum strains that carry a PfCRT variant associated with altered responsiveness to quinoline and quinoline-like antimalarial drugs. In contrast to the accumulation studies, chloroquine (CQ)-resistant parasites were observed cross-resistant to Fluo-CQ when the chemical probe was tested in various CQ-sensitive and -resistant parasite strains. NBD derivatives were found to act as redox cyclers of two essential targets, using a coupled assay based on methemoglobin and the NADPH-dependent glutathione reductase (GRs) from P. falciparum. This redox activity is proposed to contribute to the dual action of Fluo-CQ on redox equilibrium and methemoglobin reduction via PfCRT-mediated drug efflux in the cytosol and then continuous redox-dependent shuttling between food vacuole and cytosol. Taking into account these physicochemical characteristics, a model was proposed to explain Fluo-CQ antimalarial effects involving the contribution of PfCRT-mediated transport, methemoglobin reduction, hematin binding, and NBD reduction activity catalyzed by PfGR in CQ-resistant versus CQ-sensitive parasites. Therefore, introduction of NBD fluorophore in drugs is not inert and should be taken into account in drug transport and imaging studies.


Subject(s)
Chloroquine/analogs & derivatives , Membrane Transport Proteins/genetics , Plasmodium falciparum/classification , Protozoan Proteins/genetics , Antimalarials/chemistry , Antimalarials/pharmacology , Chloroquine/chemistry , Chloroquine/pharmacology , Drug Resistance , Fluorescent Dyes/chemistry , Hydrogen-Ion Concentration , Malaria, Falciparum/parasitology , Mutation , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...