Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters

Country/Region as subject
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.
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
3.
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
4.
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
5.
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
6.
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
7.
Anal Chem ; 92(8): 5765-5771, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32202408

ABSTRACT

While there is ample evidence suggesting that carriers of heterozygous hemoglobin S and C are protected from life-threatening malaria, little is known about the underlying biochemical mechanisms at the single cell level. Using nanofocused scanning X-ray fluorescence microscopy, we quantify the spatial distribution of individual elements in subcellular compartments, including Fe, S, P, Zn, and Cu, in Plasmodium falciparum-infected (P. falciparum-infected) erythrocytes carrying the wild type or variant hemoglobins. Our data indicate that heterozygous hemoglobin S and C significantly modulate biochemical reactions in parasitized erythrocytes, such as aberrant hemozoin mineralization and a delay in hemoglobin degradation. The label-free scanning X-ray fluorescence imaging has great potential to quantify the spatial distribution of elements in subcellular compartments of P. falciparum-infected erythrocytes and unravel the biochemical mechanisms underpinning disease and protective traits.


Subject(s)
Erythrocytes/metabolism , Hemoglobin C/metabolism , Hemoglobin, Sickle/metabolism , Nanotechnology , Plasmodium falciparum/metabolism , Cells, Cultured , Erythrocytes/parasitology , Hemoglobin C/analysis , Hemoglobin, Sickle/analysis , Humans , Microscopy, Fluorescence , X-Rays
9.
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
10.
Cell Microbiol ; 19(2)2017 02.
Article in English | MEDLINE | ID: mdl-27450804

ABSTRACT

During intraerythrocytic development, Plasmodium falciparum increases the ion permeability of the erythrocyte plasma membrane to an extent that jeopardizes the osmotic stability of the host cell. A previously formulated numeric model has suggested that the parasite prevents premature rupture of the host cell by consuming hemoglobin (Hb) in excess of its own anabolic needs. Here, we have tested the colloid-osmotic model on the grounds of time-resolved experimental measurements on cell surface area and volume. We have further verified whether the colloid-osmotic model can predict time-dependent volumetric changes when parasites are grown in erythrocytes containing the hemoglobin variants S or C. A good agreement between model-predicted and empirical data on both infected erythrocyte and intracellular parasite volume was found for parasitized HbAA and HbAC erythrocytes. However, a delayed induction of the new permeation pathways needed to be taken into consideration for the latter case. For parasitized HbAS erythrocyte, volumes diverged from model predictions, and infected erythrocytes showed excessive vesiculation during the replication cycle. We conclude that the colloid-osmotic model provides a plausible and experimentally supported explanation of the volume expansion and osmotic stability of P. falciparum-infected erythrocytes. The contribution of vesiculation to the malaria-protective function of hemoglobin S is discussed.


Subject(s)
Cell Membrane/physiology , Erythrocytes/cytology , Erythrocytes/parasitology , Hemoglobinopathies/pathology , Host-Pathogen Interactions , Permeability , Plasmodium falciparum/pathogenicity , Cell Shape , Cell Size , Models, Theoretical , Time Factors
11.
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
12.
Biophys J ; 112(9): 1908-1919, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28494961

ABSTRACT

To avoid clearance by the spleen, red blood cells infected with the human malaria parasite Plasmodium falciparum (iRBCs) adhere to the vascular endothelium through adhesive protrusions called "knobs" that the parasite induces on the surface of the host cell. However, the detailed relation between the developing knob structure and the resulting movement in shear flow is not known. Using flow chamber experiments on endothelial monolayers and tracking of the parasite inside the infected host cell, we find that trophozoites (intermediate-stage iRBCs) tend to flip due to their biconcave shape, whereas schizonts (late-stage iRBCs) tend to roll due to their almost spherical shape. We then use adhesive dynamics simulations for spherical cells to predict the effects of knob density and receptor multiplicity per knob on rolling adhesion of schizonts. We find that rolling adhesion requires a homogeneous coverage of the cell surface by knobs and that rolling adhesion becomes more stable and slower for higher knob density. Our experimental data suggest that schizonts are at the border between transient and stable rolling adhesion. They also allow us to establish an estimate for the molecular parameters for schizont adhesion to the vascular endothelium and to predict bond dynamics in the contact region.


Subject(s)
Cell Adhesion/physiology , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/parasitology , Erythrocytes/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/blood , Cells, Cultured , Computer Simulation , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Erythrocyte Membrane/pathology , Erythrocytes/pathology , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Models, Cardiovascular , Motion , Plasmodium falciparum , Regional Blood Flow/physiology
13.
Mol Microbiol ; 101(1): 1-11, 2016 07.
Article in English | MEDLINE | ID: mdl-26996123

ABSTRACT

The malaria parasite Plasmodium falciparum exports numerous proteins to its chosen host cell, the mature human erythrocyte. Many of these proteins are important for parasite survival. To reach the host cell, parasites must cross multiple membrane barriers and then furthermore be targeted to their correct sub-cellular localisation. This novel transport pathway has received much research attention in the past decades, especially as many of the mechanisms are expected to be parasite-specific and thus potential targets for drug development. In this article we summarize some of the most recent advances in this field, and highlight areas in which further research is needed.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/blood , Plasmodium falciparum/metabolism , Erythrocyte Membrane/metabolism , Host-Parasite Interactions/physiology , Humans , Malaria, Falciparum/parasitology , Plasmodium falciparum/parasitology , Protein Transport , Protozoan Proteins/blood
14.
Blood ; 125(2): 383-91, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25352129

ABSTRACT

Infections with the human malaria parasite Plasmodium falciparum during pregnancy can lead to severe complications for both mother and child, resulting from the cytoadhesion of parasitized erythrocytes in the intervillous space of the placenta. Cytoadherence is conferred by the specific interaction of the parasite-encoded adhesin VAR2CSA with chondroitin-4-sulfate (CSA) present on placental proteoglycans. CSA presented elsewhere in the microvasculature does not afford VAR2CSA-mediated cytoadhesion of parasitized erythrocytes. To address the placenta-specific binding tropism, we investigated the effect of the receptor/ligand arrangement on cytoadhesion, using artificial membranes with different CSA spacing intervals. We found that cytoadhesion is strongly dependent on the CSA distance, with half-maximal adhesion occurring at a CSA distance of 9 ± 1 nm at all hydrodynamic conditions. Moreover, binding to CSA was cooperative and shear stress induced. These findings suggest that the CSA density, together with allosteric effects in VAR2CSA, aid in discriminating between different CSA milieus.


Subject(s)
Cell Adhesion/physiology , Chondroitin Sulfates/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Pregnancy Complications, Parasitic/parasitology , Erythrocytes/metabolism , Female , Humans , In Vitro Techniques , Lipid Bilayers/metabolism , Microscopy, Atomic Force , Pregnancy
15.
PLoS Genet ; 10(5): e1004382, 2014 May.
Article in English | MEDLINE | ID: mdl-24830312

ABSTRACT

The emerging resistance to quinine jeopardizes the efficacy of a drug that has been used in the treatment of malaria for several centuries. To identify factors contributing to differential quinine responses in the human malaria parasite Plasmodium falciparum, we have conducted comparative quantitative trait locus analyses on the susceptibility to quinine and also its stereoisomer quinidine, and on the initial and steady-state intracellular drug accumulation levels in the F1 progeny of a genetic cross. These data, together with genetic screens of field isolates and laboratory strains associated differential quinine and quinidine responses with mutated pfcrt, a segment on chromosome 13, and a novel candidate gene, termed MAL7P1.19 (encoding a HECT ubiquitin ligase). Despite a strong likelihood of association, episomal transfections demonstrated a role for the HECT ubiquitin-protein ligase in quinine and quinidine sensitivity in only a subset of genetic backgrounds, and here the changes in IC50 values were moderate (approximately 2-fold). These data show that quinine responsiveness is a complex genetic trait with multiple alleles playing a role and that more experiments are needed to unravel the role of the contributing factors.


Subject(s)
Plasmodium falciparum/drug effects , Quinidine/pharmacology , Quinine/pharmacology , Ubiquitin-Protein Ligases/genetics , Animals , Chromosome Mapping , Endoplasmic Reticulum/enzymology , Golgi Apparatus/enzymology , Plasmodium falciparum/enzymology , Polymorphism, Genetic , Quantitative Trait Loci
16.
Proc Natl Acad Sci U S A ; 111(17): E1759-67, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24728833

ABSTRACT

Mutations in the chloroquine resistance transporter (PfCRT) are the primary determinant of chloroquine (CQ) resistance in the malaria parasite Plasmodium falciparum. A number of distinct PfCRT haplotypes, containing between 4 and 10 mutations, have given rise to CQ resistance in different parts of the world. Here we present a detailed molecular analysis of the number of mutations (and the order of addition) required to confer CQ transport activity upon the PfCRT as well as a kinetic characterization of diverse forms of PfCRT. We measured the ability of more than 100 variants of PfCRT to transport CQ when expressed at the surface of Xenopus laevis oocytes. Multiple mutational pathways led to saturable CQ transport via PfCRT, but these could be separated into two main lineages. Moreover, the attainment of full activity followed a rigid process in which mutations had to be added in a specific order to avoid reductions in CQ transport activity. A minimum of two mutations sufficed for (low) CQ transport activity, and as few as four conferred full activity. The finding that diverse PfCRT variants are all limited in their capacity to transport CQ suggests that resistance could be overcome by reoptimizing the CQ dosage.


Subject(s)
Chloroquine/metabolism , Drug Resistance , Malaria, Falciparum/metabolism , Membrane Transport Proteins/genetics , Mutation/genetics , Parasites/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Amino Acid Sequence , Animals , Biological Transport , Haplotypes , Kinetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oocytes , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transfection , Xenopus laevis
17.
Molecules ; 22(1)2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28106855

ABSTRACT

With the aim of increasing the structural diversity on the early antimalarial drug plasmodione, an efficient and versatile procedure to prepare a series of biaryl- and N-arylalkylamines as plasmodione analogues is described. Using the naturally occurring and commercially available menadione as starting material, a 2-step sequence using a Kochi-Anderson reaction and subsequent Pd-catalyzed Suzuki-Miyaura coupling was developed to prepare three representative biphenyl derivatives in good yields for antimalarial evaluation. In addition, synthetic methodologies to afford 3-benzylmenadione derivatives bearing a terminal -N(Me)2 or -N(Et)2 in different positions (ortho, meta and para) on the aryl ring of the benzylic chain of plasmodione were investigated through reductive amination was used as the optimal route to prepare these protonable N-arylalkylamine privileged scaffolds. The antimalarial activities were evaluated and discussed in light of their physicochemical properties. Among the newly synthesized compounds, the para-position of the substituent remains the most favourable position on the benzyl chain and the carbamate -NHBoc was found active both in vitro (42 nM versus 29 nM for plasmodione) and in vivo in Plasmodium berghei-infected mice. The measured acido-basic features of these new molecules support the cytosol-food vacuole shuttling properties of non-protonable plasmodione derivatives essential for redox-cycling. These findings may be useful in antimalarial drug optimization.


Subject(s)
Amines/administration & dosage , Amines/chemical synthesis , Antimalarials/administration & dosage , Antimalarials/chemical synthesis , Malaria/drug therapy , Amines/chemistry , Amines/pharmacology , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Combinatorial Chemistry Techniques , Mice , Molecular Structure , Oxidation-Reduction , Plasmodium berghei/drug effects , Structure-Activity Relationship , Vitamin K 3/analogs & derivatives
18.
Mol Microbiol ; 97(2): 381-95, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25898991

ABSTRACT

The widespread use of chloroquine to treat Plasmodium falciparum infections has resulted in the selection and dissemination of variant haplotypes of the primary resistance determinant PfCRT. These haplotypes have encountered drug pressure and within-host competition with wild-type drug-sensitive parasites. To examine these selective forces in vitro, we genetically engineered P. falciparum to express geographically diverse PfCRT haplotypes. Variant alleles from the Philippines (PH1 and PH2, which differ solely by the C72S mutation) both conferred a moderate gain of chloroquine resistance and a reduction in growth rates in vitro. Of the two, PH2 showed higher IC50 values, contrasting with reduced growth. Furthermore, a highly mutated pfcrt allele from Cambodia (Cam734) conferred moderate chloroquine resistance and enhanced growth rates, when tested against wild-type pfcrt in co-culture competition assays. These three alleles mediated cross-resistance to amodiaquine, an antimalarial drug widely used in Africa. Each allele, along with the globally prevalent Dd2 and 7G8 alleles, rendered parasites more susceptible to lumefantrine, the partner drug used in the leading first-line artemisinin-based combination therapy. These data reveal ongoing region-specific evolution of PfCRT that impacts drug susceptibility and relative fitness in settings of mixed infections, and raise important considerations about optimal agents to treat chloroquine-resistant malaria.


Subject(s)
Membrane Transport Proteins/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Chloroquine , Drug Resistance , Erythrocytes/parasitology , Gene Frequency , Haplotypes , Humans , Malaria, Falciparum/parasitology , Membrane Transport Proteins/metabolism , Mutation , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism
19.
Antimicrob Agents Chemother ; 60(9): 5146-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27297478

ABSTRACT

Previously, we presented the chemical design of a promising series of antimalarial agents, 3-[substituted-benzyl]-menadiones, with potent in vitro and in vivo activities. Ongoing studies on the mode of action of antimalarial 3-[substituted-benzyl]-menadiones revealed that these agents disturb the redox balance of the parasitized erythrocyte by acting as redox cyclers-a strategy that is broadly recognized for the development of new antimalarial agents. Here we report a detailed parasitological characterization of the in vitro activity profile of the lead compound 3-[4-(trifluoromethyl)benzyl]-menadione 1c (henceforth called plasmodione) against intraerythrocytic stages of the human malaria parasite Plasmodium falciparum We show that plasmodione acts rapidly against asexual blood stages, thereby disrupting the clinically relevant intraerythrocytic life cycle of the parasite, and furthermore has potent activity against early gametocytes. The lead's antiplasmodial activity was unaffected by the most common mechanisms of resistance to clinically used antimalarials. Moreover, plasmodione has a low potential to induce drug resistance and a high killing speed, as observed by culturing parasites under continuous drug pressure. Drug interactions with licensed antimalarial drugs were also established using the fixed-ratio isobologram method. Initial toxicological profiling suggests that plasmodione is a safe agent for possible human use. Our studies identify plasmodione as a promising antimalarial lead compound and strongly support the future development of redox-active benzylmenadiones as antimalarial agents.


Subject(s)
Antimalarials/pharmacology , Gametogenesis/drug effects , Life Cycle Stages/drug effects , Naphthoquinones/pharmacology , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Artemisinins/pharmacology , Atovaquone/pharmacology , Drug Interactions , Drug Resistance/drug effects , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Inhibitory Concentration 50 , Methylene Blue/pharmacology , Naphthoquinones/chemical synthesis , Plasmodium falciparum/growth & development
20.
Blood ; 124(23): 3459-68, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25139348

ABSTRACT

Following invasion of human red blood cells (RBCs) by the malaria parasite, Plasmodium falciparum, a remarkable process of remodeling occurs in the host cell mediated by trafficking of several hundred effector proteins to the RBC compartment. The exported virulence protein, P falciparum erythrocyte membrane protein 1 (PfEMP1), is responsible for cytoadherence of infected cells to host endothelial receptors. Maurer clefts are organelles essential for protein trafficking, sorting, and assembly of protein complexes. Here we demonstrate that disruption of PfEMP1 trafficking protein 1 (PfPTP1) function leads to severe alterations in the architecture of Maurer's clefts. Furthermore, 2 major surface antigen families, PfEMP1 and STEVOR, are no longer displayed on the host cell surface leading to ablation of cytoadherence to host receptors. PfPTP1 functions in a large complex of proteins and is required for linking of Maurer's clefts to the host actin cytoskeleton.


Subject(s)
Actin Cytoskeleton/metabolism , Antigens, Protozoan/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Virulence Factors/metabolism , Actins/metabolism , Cells, Cultured , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/parasitology , Erythrocytes/metabolism , Host-Parasite Interactions , Humans , Malaria, Falciparum/blood , Plasmodium falciparum/pathogenicity , Protein Transport , Transport Vesicles/metabolism , Transport Vesicles/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL