Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 20(5): e3001616, 2022 05.
Article in English | MEDLINE | ID: mdl-35507548

ABSTRACT

Polymorphisms in the Plasmodium falciparum multidrug resistance protein 1 (pfmdr1) gene and the Plasmodium falciparum chloroquine resistance transporter (pfcrt) gene alter the malaria parasite's susceptibility to most of the current antimalarial drugs. However, the precise mechanisms by which PfMDR1 contributes to multidrug resistance have not yet been fully elucidated, nor is it understood why polymorphisms in pfmdr1 and pfcrt that cause chloroquine resistance simultaneously increase the parasite's susceptibility to lumefantrine and mefloquine-a phenomenon known as collateral drug sensitivity. Here, we present a robust expression system for PfMDR1 in Xenopus oocytes that enables direct and high-resolution biochemical characterizations of the protein. We show that wild-type PfMDR1 transports diverse pharmacons, including lumefantrine, mefloquine, dihydroartemisinin, piperaquine, amodiaquine, methylene blue, and chloroquine (but not the antiviral drug amantadine). Field-derived mutant isoforms of PfMDR1 differ from the wild-type protein, and each other, in their capacities to transport these drugs, indicating that PfMDR1-induced changes in the distribution of drugs between the parasite's digestive vacuole (DV) and the cytosol are a key driver of both antimalarial resistance and the variability between multidrug resistance phenotypes. Of note, the PfMDR1 isoforms prevalent in chloroquine-resistant isolates exhibit reduced capacities for chloroquine, lumefantrine, and mefloquine transport. We observe the opposite relationship between chloroquine resistance-conferring mutations in PfCRT and drug transport activity. Using our established assays for characterizing PfCRT in the Xenopus oocyte system and in live parasite assays, we demonstrate that these PfCRT isoforms transport all 3 drugs, whereas wild-type PfCRT does not. We present a mechanistic model for collateral drug sensitivity in which mutant isoforms of PfMDR1 and PfCRT cause chloroquine, lumefantrine, and mefloquine to remain in the cytosol instead of sequestering within the DV. This change in drug distribution increases the access of lumefantrine and mefloquine to their primary targets (thought to be located outside of the DV), while simultaneously decreasing chloroquine's access to its target within the DV. The mechanistic insights presented here provide a basis for developing approaches that extend the useful life span of antimalarials by exploiting the opposing selection forces they exert upon PfCRT and PfMDR1.


Subject(s)
Antimalarials , Malaria, Falciparum , Parasites , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Chloroquine/metabolism , Chloroquine/pharmacology , Chloroquine/therapeutic use , Drug Resistance/genetics , Drug Resistance, Multiple , Lumefantrine/pharmacology , Lumefantrine/therapeutic use , Malaria, Falciparum/parasitology , Mefloquine/metabolism , Mefloquine/pharmacology , Mefloquine/therapeutic use , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/therapeutic use , Parasites/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Isoforms/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
2.
Sci Rep ; 11(1): 6787, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33762657

ABSTRACT

Toxoplasma gondii and Plasmodium falciparum parasites both extrude L-lactate, a byproduct of glycolysis. The P. falciparum Formate Nitrite Transporter, PfFNT, mediates L-lactate transport across the plasma membrane of P. falciparum parasites and has been validated as a drug target. The T. gondii genome encodes three FNTs that have been shown to transport L-lactate, and which are proposed to be the targets of several inhibitors of T. gondii proliferation. Here, we show that each of the TgFNTs localize to the T. gondii plasma membrane and are capable of transporting L-lactate across it, with TgFNT1 making the primary contribution to L-lactate transport during the disease-causing lytic cycle of the parasite. We use the Xenopus oocyte expression system to provide direct measurements of L-lactate transport via TgFNT1. We undertake a genetic analysis of the importance of the tgfnt genes for parasite proliferation, and demonstrate that all three tgfnt genes can be disrupted individually and together without affecting the lytic cycle under in vitro culture conditions. Together, our experiments identify the major lactate transporter in the disease causing stage of T. gondii, and reveal that this transporter is not required for parasite proliferation, indicating that TgFNTs are unlikely to be targets for anti-Toxoplasma drugs.


Subject(s)
Monocarboxylic Acid Transporters/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Animals , Cell Membrane/metabolism , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/genetics , Oocytes/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protozoan Proteins/genetics , Toxoplasma/growth & development , Xenopus/growth & development
3.
Eur J Med Chem ; 215: 113227, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33601312

ABSTRACT

Due to the surge in resistance to common therapies, malaria remains a significant concern to human health worldwide. In chloroquine (CQ)-resistant (CQ-R) strains of Plasmodium falciparum, CQ and related drugs are effluxed from the parasite's digestive vacuole (DV). This process is mediated by mutant isoforms of a protein called CQ resistance transporter (PfCRT). CQ-R strains can be partially re-sensitized to CQ by verapamil (VP), primaquine (PQ) and other compounds, and this has been shown to be due to the ability of these molecules to inhibit drug transport via PfCRT. We have previously developed a series of clotrimazole (CLT)-based antimalarial agents that possess inhibitory activity against PfCRT (4a,b). In our endeavor to develop novel PfCRT inhibitors, and to perform a structure-activity relationship analysis, we synthesized a new library of analogues. When the benzhydryl system was linked to a 4-aminoquinoline group (5a-f) the resulting compounds exhibited good cytotoxicity against both CQ-R and CQ-S strains of P. falciparum. The most potent inhibitory activity against the PfCRT-mediated transport of CQ was obtained with compound 5k. When compared to the reference compound, benzhydryl analogues of PQ (5i,j) showed a similar activity against blood-stage parasites, and a stronger in vitro potency against liver-stage parasites. Unfortunately, in the in vivo transmission blocking assays, 5i,j were inactive against gametocytes.


Subject(s)
Antimalarials/pharmacology , Benzhydryl Compounds/pharmacology , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Animals , Anopheles , Antimalarials/chemical synthesis , Benzhydryl Compounds/chemical synthesis , Chloroquine/pharmacology , Drug Design , Drug Resistance, Microbial/drug effects , Female , Hep G2 Cells , Humans , Membrane Transport Proteins , Mice , Mice, Inbred BALB C , Molecular Structure , NIH 3T3 Cells , Parasitic Sensitivity Tests , Protein Isoforms/antagonists & inhibitors , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Xenopus
4.
PLoS Comput Biol ; 16(9): e1008243, 2020 09.
Article in English | MEDLINE | ID: mdl-32991585

ABSTRACT

Understanding evolution requires detailed knowledge of genotype-phenotype maps; however, it can be a herculean task to measure every phenotype in a combinatorial map. We have developed a computational strategy to predict the missing phenotypes from an incomplete, combinatorial genotype-phenotype map. As a test case, we used an incomplete genotype-phenotype dataset previously generated for the malaria parasite's 'chloroquine resistance transporter' (PfCRT). Wild-type PfCRT (PfCRT3D7) lacks significant chloroquine (CQ) transport activity, but the introduction of the eight mutations present in the 'Dd2' isoform of PfCRT (PfCRTDd2) enables the protein to transport CQ away from its site of antimalarial action. This gain of a transport function imparts CQ resistance to the parasite. A combinatorial map between PfCRT3D7 and PfCRTDd2 consists of 256 genotypes, of which only 52 have had their CQ transport activities measured through expression in the Xenopus laevis oocyte. We trained a statistical model with these 52 measurements to infer the CQ transport activity for the remaining 204 combinatorial genotypes between PfCRT3D7 and PfCRTDd2. Our best-performing model incorporated a binary classifier, a nonlinear scale, and additive effects for each mutation. The addition of specific pairwise- and high-order-epistatic coefficients decreased the predictive power of the model. We evaluated our predictions by experimentally measuring the CQ transport activities of 24 additional PfCRT genotypes. The R2 value between our predicted and newly-measured phenotypes was 0.90. We then used the model to probe the accessibility of evolutionary trajectories through the map. Approximately 1% of the possible trajectories between PfCRT3D7 and PfCRTDd2 are accessible; however, none of the trajectories entailed eight successive increases in CQ transport activity. These results demonstrate that phenotypes can be inferred with known uncertainty from a partial genotype-phenotype dataset. We also validated our approach against a collection of previously published genotype-phenotype maps. The model therefore appears general and should be applicable to a large number of genotype-phenotype maps.


Subject(s)
Genotype , Phenotype , Animals , Models, Biological , Mutation , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Uncertainty
5.
Nat Commun ; 11(1): 3922, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32764664

ABSTRACT

The Plasmodium falciparum chloroquine resistance transporter (PfCRT) is a key contributor to multidrug resistance and is also essential for the survival of the malaria parasite, yet its natural function remains unresolved. We identify host-derived peptides of 4-11 residues, varying in both charge and composition, as the substrates of PfCRT in vitro and in situ, and show that PfCRT does not mediate the non-specific transport of other metabolites and/or ions. We find that drug-resistance-conferring mutations reduce both the peptide transport capacity and substrate range of PfCRT, explaining the impaired fitness of drug-resistant parasites. Our results indicate that PfCRT transports peptides from the lumen of the parasite's digestive vacuole to the cytosol, thereby providing a source of amino acids for parasite metabolism and preventing osmotic stress of this organelle. The resolution of PfCRT's native substrates will aid the development of drugs that target PfCRT and/or restore the efficacy of existing antimalarials.


Subject(s)
Antimalarials/pharmacology , Chloroquine/pharmacology , Membrane Transport Proteins/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Biological Transport, Active , Drug Resistance/genetics , Female , Host-Parasite Interactions/genetics , Host-Parasite Interactions/physiology , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Membrane Transport Proteins/genetics , Models, Biological , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oligopeptides/metabolism , Oocytes/metabolism , Plasmodium falciparum/genetics , Protein Transport , Protozoan Proteins/genetics , Xenopus laevis
6.
Pract Radiat Oncol ; 10(4): e238-e243, 2020.
Article in English | MEDLINE | ID: mdl-31676405

ABSTRACT

PURPOSE: To compare whole-breast radiation therapy dose distributions with and without the use of breast cups to reposition the breast itself. METHODS AND MATERIALS: A fitted plastic breast cup repositions the breast on the anterior chest wall, reducing the lateral fall and ptosis of the breast. Eighteen patients requiring breast cups were identified on an initial simulation computed tomography scan (without breast cup); subsequently, a second scan with the breast cup fitted was performed. An optimal treatment plan was then created on both scans with the differences in radiation dose to underlying organs compared. In vivo measurements were performed to assess any possible bolusing effect on skin dose owing to the breast cup. RESULTS: Breast cups significantly reduce dose to both lung and heart for all left-sided cases. All lung dose metrics decreased for all right-sided cases. Right-sided heart dose metrics did not significantly decrease with the use of breast cups; however, heart mean dose for these cases was all under 100 cGy. A 16% increase in skin surface dose was observed in an anthropomorphic phantom when using a breast cup. CONCLUSIONS: Use of the breast cup in breast radiation therapy decreases dose to underlying organs at risk and is cost-effective. It can be easily integrated with deep inspiration breath hold, intensity modulated radiation therapy, and volumetric-modulated arc therapy techniques. With use of the device, a supine patient position can be maintained, meaning nodal regions can be treated mono-isocentrically.


Subject(s)
Breast Neoplasms/radiotherapy , Breast/radiation effects , Female , Humans
7.
Biol Rev Camb Philos Soc ; 95(2): 305-332, 2020 04.
Article in English | MEDLINE | ID: mdl-31701663

ABSTRACT

Membrane transport proteins, also known as transporters, control the movement of ions, nutrients, metabolites, and waste products across the membranes of a cell and are central to its biology. Proteins of this type also serve as drug targets and are key players in the phenomenon of drug resistance. The malaria parasite has a relatively reduced transportome, with only approximately 2.5% of its genes encoding transporters. Even so, assigning functions and physiological roles to these proteins, and ascertaining their contributions to drug action and drug resistance, has been very challenging. This review presents a detailed critique and synthesis of the disruption phenotypes, protein subcellular localisations, protein functions (observed or predicted), and links to antimalarial drug resistance for each of the parasite's transporter genes. The breadth and depth of the gene disruption data are particularly impressive, with at least one phenotype determined in the parasite's asexual blood stage for each transporter gene, and multiple phenotypes available for 76% of the genes. Analysis of the curated data set revealed there to be relatively little redundancy in the Plasmodium transportome; almost two-thirds of the parasite's transporter genes are essential or required for normal growth in the asexual blood stage of the parasite, and this proportion increased to 78% when the disruption phenotypes available for the other parasite life stages were included in the analysis. These observations, together with the finding that 22% of the transportome is implicated in the parasite's resistance to existing antimalarials and/or drugs within the development pipeline, indicate that transporters are likely to serve, or are already serving, as drug targets. Integration of the different biological and bioinformatic data sets also enabled the selection of candidates for transport processes known to be essential for parasite survival, but for which the underlying proteins have thus far remained undiscovered. These include potential transporters of pantothenate, isoleucine, or isopentenyl diphosphate, as well as putative anion-selective channels that may serve as the pore component of the parasite's 'new permeation pathways'. Other novel insights into the parasite's biology included the identification of transporters for the potential development of antimalarial treatments, transmission-blocking drugs, prophylactics, and genetically attenuated vaccines. The syntheses presented herein set a foundation for elucidating the functions and physiological roles of key members of the Plasmodium transportome and, ultimately, to explore and realise their potential as therapeutic targets.


Subject(s)
Gene Expression Regulation/physiology , Plasmodium/physiology , Biological Transport/genetics , Biological Transport/physiology , Genome, Protozoan
8.
Front Physiol ; 10: 1375, 2019.
Article in English | MEDLINE | ID: mdl-31736791

ABSTRACT

Dopamine is an important neuromodulator involved in reward-processing, movement control, motivational responses, and other aspects of behavior in most animals. In honey bees (Apis mellifera), the dopaminergic system has been implicated in an elaborate pheromonal communication network between individuals and in the differentiation of females into reproductive (queen) and sterile (worker) castes. Here we have identified and characterized a honey bee dopamine transporter (AmDAT) and a splice variant lacking exon 3 (AmDATΔex3). Both transcripts are present in the adult brain and antennae as well as at lower levels within larvae and ovaries. When expressed separately in the Xenopus oocyte system, AmDAT localizes to the oocyte surface whereas the splice variant is retained at an internal membrane. Oocytes expressing AmDAT exhibit a 12-fold increase in the uptake of [3H]dopamine relative to non-injected oocytes, whereas the AmDATΔex3-expressing oocytes show no change in [3H]dopamine transport. Electrophysiological measurements of AmDAT activity revealed it to be a high-affinity, low-capacity transporter of dopamine. The transporter also recognizes noradrenaline as a major substrate and tyramine as a minor substrate, but does not transport octopamine, L-Dopa, or serotonin. Dopamine transport via AmDAT is inhibited by cocaine in a reversible manner, but is unaffected by octopamine. Co-expression of AmDAT and AmDATΔex3 in oocytes results in a substantial reduction in AmDAT-mediated transport, which was also detected as a significant decrease in the level of AmDAT protein. This down-regulatory effect is not attributable to competition with AmDATΔex3 for ER ribosomes, nor to a general inhibition of the oocyte's translational machinery. In vivo, the expression of both transcripts shows a high level of inter-individual variability. Gene-focused, ultra-deep amplicon sequencing detected methylation of the amdat locus at ten 5'-C-phosphate-G-3' dinucleotides (CpGs), but only in 5-10% of all reads in whole brains or antennae. These observations, together with the localization of the amdat transcript to a few clusters of dopaminergic neurons, imply that amdat methylation is positively linked to its transcription. Our findings suggest that multiple cellular mechanisms, including gene splicing and epigenomic communication systems, may be adopted to increase the potential of a conserved gene to contribute to lineage-specific behavioral outcomes.

9.
J Med Imaging Radiat Oncol ; 62(6): 847-853, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30183132

ABSTRACT

INTRODUCTION: There has been no systematic attempt to enhance leadership capacity within radiation oncology as an integrated component of training. This pilot study examines an intervention to introduce basics of leadership learning to radiation oncology trainees. METHODS: A case-based learning tool was designed for delivery via trainees' personal electronic devices. Eight typical workplace case scenarios representing leadership challenges were followed by multiple choice questions, key learning points and hyperlinks to relevant resources. Cases were automatically sent every few days over 4 weeks and participants' responses anonymously collated by the delivery platform (QStream). In addition, an online survey was sent at completion of the program to capture trainees' perspectives on the utility of this tool. RESULTS: Thirty-seven of 45 (82%) trainees participated: 21 females and 16 males. Twenty-six of 37 (70%) starting the program completed it. Sixteen (62% of 'completers') responded to the post-program survey. Fourteen of 16 (87.5%) agreed to the program and helped them identify ways they were already exhibiting leadership. Eleven of 16 (68.8%) agreed they had acquired knowledge that could assist them in being better leaders. Fifteen of 16 said the program made them consider future leadership possibilities in radiation oncology. Fourteen of 15 enjoyed the digital format. Most suggestions for improvement linked to a desire for more interactivity in learning these skills. CONCLUSION: Piloting an online tool designed to introduce foundation leadership concepts to radiation oncology trainees has provided useful feedback to guide further development in this area. Although this method had high feasibility, it revealed the need for additional interactive methods for leadership learning.


Subject(s)
Computer-Assisted Instruction , Education, Medical, Graduate/organization & administration , Leadership , Radiation Oncologists , Radiation Oncology/education , Adult , Career Choice , Career Mobility , Clinical Competence , Female , Humans , Male , New South Wales , Pilot Projects
10.
Curr Opin Pharmacol ; 42: 71-80, 2018 10.
Article in English | MEDLINE | ID: mdl-30142480

ABSTRACT

The deployment of artemisinin-based combination therapies (ACTs) has been, and continues to be, integral to reducing the number of malaria cases and deaths. However, their efficacy is being increasingly jeopardized by the emergence and spread of parasites that are resistant (or partially resistant) to the artemisinin derivatives and to their partner drugs, with the efficacy of the latter being especially crucial for treatment success. A detailed understanding of the genetic determinants of resistance to the ACT partner drugs, and the mechanisms by which they mediate resistance, is required for the surveillance of molecular markers and to optimize the efficacy and lifespan of the partner drugs through resistance management strategies. We summarize new insights into the molecular basis of parasite resistance to the ACTs, such as recently-uncovered determinants of parasite susceptibility to the artemisinin derivatives, piperaquine, lumefantrine, and mefloquine, and outline the mechanisms through which polymorphisms in these determinants may be conferring resistance.


Subject(s)
Artemisinins/pharmacology , Artemisinins/therapeutic use , Drug Resistance/drug effects , Malaria/drug therapy , Parasites/drug effects , Animals , Humans
11.
Cell ; 170(2): 260-272.e8, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28708996

ABSTRACT

The genomes of malaria parasites contain many genes of unknown function. To assist drug development through the identification of essential genes and pathways, we have measured competitive growth rates in mice of 2,578 barcoded Plasmodium berghei knockout mutants, representing >50% of the genome, and created a phenotype database. At a single stage of its complex life cycle, P. berghei requires two-thirds of genes for optimal growth, the highest proportion reported from any organism and a probable consequence of functional optimization necessitated by genomic reductions during the evolution of parasitism. In contrast, extreme functional redundancy has evolved among expanded gene families operating at the parasite-host interface. The level of genetic redundancy in a single-celled organism may thus reflect the degree of environmental variation it experiences. In the case of Plasmodium parasites, this helps rationalize both the relative successes of drugs and the greater difficulty of making an effective vaccine.


Subject(s)
Genome, Protozoan , Plasmodium berghei/growth & development , Plasmodium berghei/genetics , Animals , Biological Evolution , Female , Gene Knockout Techniques , Genes, Essential , Host-Parasite Interactions , Metabolic Networks and Pathways , Mice , Mice, Inbred BALB C , Plasmodium berghei/metabolism , Saccharomyces cerevisiae/genetics , Toxoplasma/genetics , Trypanosoma brucei brucei/genetics
12.
PLoS Pathog ; 13(2): e1006180, 2017 02.
Article in English | MEDLINE | ID: mdl-28178359

ABSTRACT

In this study the 'Malaria Box' chemical library comprising 400 compounds with antiplasmodial activity was screened for compounds that perturb the internal pH of the malaria parasite, Plasmodium falciparum. Fifteen compounds induced an acidification of the parasite cytosol. Two of these did so by inhibiting the parasite's formate nitrite transporter (PfFNT), which mediates the H+-coupled efflux from the parasite of lactate generated by glycolysis. Both compounds were shown to inhibit lactate transport across the parasite plasma membrane, and the transport of lactate by PfFNT expressed in Xenopus laevis oocytes. PfFNT inhibition caused accumulation of lactate in parasitised erythrocytes, and swelling of both the parasite and parasitised erythrocyte. Long-term exposure of parasites to one of the inhibitors gave rise to resistant parasites with a mutant form of PfFNT that showed reduced inhibitor sensitivity. This study provides the first evidence that PfFNT is a druggable antimalarial target.


Subject(s)
Antimalarials/pharmacology , Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Monocarboxylic Acid Transporters/drug effects , Plasmodium falciparum/drug effects , Animals , Antimalarials/chemistry , Biological Transport/drug effects , Chromatography, Liquid , Drug Evaluation, Preclinical , Humans , Malaria, Falciparum/parasitology , Mass Spectrometry , Plasmodium falciparum/metabolism , Plasmodium falciparum/parasitology , Protozoan Proteins/metabolism , Xenopus laevis
13.
PLoS Pathog ; 12(7): e1005725, 2016 07.
Article in English | MEDLINE | ID: mdl-27441371

ABSTRACT

Mutations in the Plasmodium falciparum 'chloroquine resistance transporter' (PfCRT) confer resistance to chloroquine (CQ) and related antimalarials by enabling the protein to transport these drugs away from their targets within the parasite's digestive vacuole (DV). However, CQ resistance-conferring isoforms of PfCRT (PfCRTCQR) also render the parasite hypersensitive to a subset of structurally-diverse pharmacons. Moreover, mutations in PfCRTCQR that suppress the parasite's hypersensitivity to these molecules simultaneously reinstate its sensitivity to CQ and related drugs. We sought to understand these phenomena by characterizing the functions of PfCRTCQR isoforms that cause the parasite to become hypersensitive to the antimalarial quinine or the antiviral amantadine. We achieved this by measuring the abilities of these proteins to transport CQ, quinine, and amantadine when expressed in Xenopus oocytes and complemented this work with assays that detect the drug transport activity of PfCRT in its native environment within the parasite. Here we describe two mechanistic explanations for PfCRT-induced drug hypersensitivity. First, we show that quinine, which normally accumulates inside the DV and therewithin exerts its antimalarial effect, binds extremely tightly to the substrate-binding site of certain isoforms of PfCRTCQR. By doing so it likely blocks the normal physiological function of the protein, which is essential for the parasite's survival, and the drug thereby gains an additional killing effect. In the second scenario, we show that although amantadine also sequesters within the DV, the parasite's hypersensitivity to this drug arises from the PfCRTCQR-mediated transport of amantadine from the DV into the cytosol, where it can better access its antimalarial target. In both cases, the mutations that suppress hypersensitivity also abrogate the ability of PfCRTCQR to transport CQ, thus explaining why rescue from hypersensitivity restores the parasite's sensitivity to this antimalarial. These insights provide a foundation for understanding clinically-relevant observations of inverse drug susceptibilities in the malaria parasite.


Subject(s)
Antimalarials/pharmacology , Drug Resistance/physiology , Malaria, Falciparum , Membrane Transport Proteins/metabolism , Plasmodium falciparum/drug effects , Protozoan Proteins/metabolism , Amantadine/metabolism , Amantadine/pharmacology , Animals , Antimalarials/metabolism , Biological Transport/physiology , Blotting, Western , Chloroquine/metabolism , Chloroquine/pharmacology , Fluorescent Antibody Technique , Humans , Mutagenesis, Site-Directed , Protein Isoforms/metabolism , Quinine/metabolism , Quinine/pharmacology , Xenopus laevis
14.
Nat Commun ; 7: 11553, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27189525

ABSTRACT

Antimalarial chemotherapy, globally reliant on artemisinin-based combination therapies (ACTs), is threatened by the spread of drug resistance in Plasmodium falciparum parasites. Here we use zinc-finger nucleases to genetically modify the multidrug resistance-1 transporter PfMDR1 at amino acids 86 and 184, and demonstrate that the widely prevalent N86Y mutation augments resistance to the ACT partner drug amodiaquine and the former first-line agent chloroquine. In contrast, N86Y increases parasite susceptibility to the partner drugs lumefantrine and mefloquine, and the active artemisinin metabolite dihydroartemisinin. The PfMDR1 N86 plus Y184F isoform moderately reduces piperaquine potency in strains expressing an Asian/African variant of the chloroquine resistance transporter PfCRT. Mutations in both digestive vacuole-resident transporters are thought to differentially regulate ACT drug interactions with host haem, a product of parasite-mediated haemoglobin degradation. Global mapping of these mutations illustrates where the different ACTs could be selectively deployed to optimize treatment based on regional differences in PfMDR1 haplotypes.


Subject(s)
Antimalarials , Artemisinins , Drug Resistance, Microbial/genetics , Multidrug Resistance-Associated Proteins/genetics , Plasmodium falciparum/genetics , Drug Therapy, Combination , Geography , Haplotypes , Malaria/drug therapy , Malaria/parasitology , Mutation
15.
J Infect Dis ; 213(5): 800-10, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26503982

ABSTRACT

BACKGROUND: It is becoming increasingly apparent that certain mutations in the Plasmodium falciparum chloroquine resistance transporter (PfCRT) alter the parasite's susceptibility to diverse compounds. Here we investigated the interaction of PfCRT with 3 tricyclic compounds that have been used to treat malaria (quinacrine [QC] and methylene blue [MB]) or to study P. falciparum (acridine orange [AO]). METHODS: We measured the antiplasmodial activities of QC, MB, and AO against chloroquine-resistant and chloroquine-sensitive P. falciparum and determined whether QC and AO affect the accumulation and activity of chloroquine in these parasites. We also assessed the ability of mutant (PfCRT(Dd2)) and wild-type (PfCRT(D10)) variants of the protein to transport QC, MB, and AO when expressed at the surface of Xenopus laevis oocytes. RESULTS: Chloroquine resistance-conferring isoforms of PfCRT reduced the susceptibility of the parasite to QC, MB, and AO. In chloroquine-resistant (but not chloroquine-sensitive) parasites, AO and QC increased the parasite's accumulation of, and susceptibility to, chloroquine. All 3 compounds were shown to bind to PfCRT(Dd2), and the transport of QC and MB via this protein was saturable and inhibited by the chloroquine resistance-reverser verapamil. CONCLUSIONS: Our findings reveal that the PfCRT(Dd2)-mediated transport of tricyclic antimalarials reduces the parasite's susceptibility to these drugs.


Subject(s)
Membrane Transport Proteins/metabolism , Methylene Blue/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Quinacrine/metabolism , Verapamil/pharmacology , Animals , Antimalarials/pharmacology , Biological Transport/drug effects , Drug Resistance , Gene Expression Regulation/physiology , Genetic Variation , Oocytes/metabolism , Xenopus laevis
16.
Sci Rep ; 5: 14552, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26420308

ABSTRACT

Mutations in the Plasmodium falciparum chloroquine resistance transporter, PfCRT, are the major determinant of chloroquine resistance in this lethal human malaria parasite. Here, we describe P. falciparum lines subjected to selection by amantadine or blasticidin that carry PfCRT mutations (C101F or L272F), causing the development of enlarged food vacuoles. These parasites also have increased sensitivity to chloroquine and some other quinoline antimalarials, but exhibit no or minimal change in sensitivity to artemisinins, when compared with parental strains. A transgenic parasite line expressing the L272F variant of PfCRT confirmed this increased chloroquine sensitivity and enlarged food vacuole phenotype. Furthermore, the introduction of the C101F or L272F mutation into a chloroquine-resistant variant of PfCRT reduced the ability of this protein to transport chloroquine by approximately 93 and 82%, respectively, when expressed in Xenopus oocytes. These data provide, at least in part, a mechanistic explanation for the increased sensitivity of the mutant parasite lines to chloroquine. Taken together, these findings provide new insights into PfCRT function and PfCRT-mediated drug resistance, as well as the food vacuole, which is an important target of many antimalarial drugs.


Subject(s)
Chloroquine/pharmacology , Drug Resistance/genetics , Membrane Transport Proteins/genetics , Mutation , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Protozoan Proteins/genetics , Vacuoles/metabolism , Amino Acid Substitution , Animals , Antimalarials/pharmacology , Biological Transport , Humans , Membrane Transport Proteins/chemistry , Oocytes/metabolism , Parasitic Sensitivity Tests , Protozoan Proteins/chemistry , Xenopus laevis
17.
Nat Commun ; 6: 6721, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25823844

ABSTRACT

The intraerythrocytic malaria parasite relies primarily on glycolysis to fuel its rapid growth and reproduction. The major byproduct of this metabolism, lactic acid, is extruded into the external medium. In this study, we show that the human malaria parasite Plasmodium falciparum expresses at its surface a member of the microbial formate-nitrite transporter family (PfFNT), which, when expressed in Xenopus laevis oocytes, transports both formate and lactate. The transport characteristics of PfFNT in oocytes (pH-dependence, inhibitor-sensitivity and kinetics) are similar to those of the transport of lactate and formate across the plasma membrane of mature asexual-stage P. falciparum trophozoites, consistent with PfFNT playing a major role in the efflux of lactate and hence in the energy metabolism of the intraerythrocytic parasite.


Subject(s)
Formates/metabolism , Lactic Acid/metabolism , Membrane Transport Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Glycolysis , Oocytes/metabolism , Xenopus laevis
18.
J Biol Chem ; 289(52): 36336-51, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25378409

ABSTRACT

Mutations in the "chloroquine resistance transporter" (PfCRT) are a major determinant of drug resistance in the malaria parasite Plasmodium falciparum. We have previously shown that mutant PfCRT transports the antimalarial drug chloroquine away from its target, whereas the wild-type form of PfCRT does not. However, little is understood about the transport of other drugs via PfCRT or the mechanism by which PfCRT recognizes different substrates. Here we show that mutant PfCRT also transports quinine, quinidine, and verapamil, indicating that the protein behaves as a multidrug resistance carrier. Detailed kinetic analyses revealed that chloroquine and quinine compete for transport via PfCRT in a manner that is consistent with mixed-type inhibition. Moreover, our analyses suggest that PfCRT accepts chloroquine and quinine at distinct but antagonistically interacting sites. We also found verapamil to be a partial mixed-type inhibitor of chloroquine transport via PfCRT, further supporting the idea that PfCRT possesses multiple substrate-binding sites. Our findings provide new mechanistic insights into the workings of PfCRT, which could be exploited to design potent inhibitors of this key mediator of drug resistance.


Subject(s)
Antimalarials/metabolism , Membrane Transport Proteins/physiology , Plasmodium falciparum/metabolism , Protozoan Proteins/physiology , Animals , Antimalarials/pharmacology , Binding Sites , Binding, Competitive , Biological Transport , Cells, Cultured , Chloroquine/metabolism , Chloroquine/pharmacology , Drug Resistance , Female , Hydrogen-Ion Concentration , Kinetics , Protozoan Proteins/antagonists & inhibitors , Quinidine/metabolism , Quinine/metabolism , Verapamil/metabolism , Verapamil/pharmacology , Xenopus laevis
19.
Biosci Rep ; 34(6): e00150, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25405893

ABSTRACT

Although efforts to understand the basis for inter-strain phenotypic variation in the most virulent malaria species, Plasmodium falciparum, have benefited from advances in genomic technologies, there have to date been few metabolomic studies of this parasite. Using 1H-NMR spectroscopy, we have compared the metabolite profiles of red blood cells infected with different P. falciparum strains. These included both chloroquine-sensitive and chloroquine-resistant strains, as well as transfectant lines engineered to express different isoforms of the chloroquine-resistance-conferring pfcrt (P. falciparum chloroquine resistance transporter). Our analyses revealed strain-specific differences in a range of metabolites. There was marked variation in the levels of the membrane precursors choline and phosphocholine, with some strains having >30-fold higher choline levels and >5-fold higher phosphocholine levels than others. Chloroquine-resistant strains showed elevated levels of a number of amino acids relative to chloroquine-sensitive strains, including an approximately 2-fold increase in aspartate levels. The elevation in amino acid levels was attributable to mutations in pfcrt. Pfcrt-linked differences in amino acid abundance were confirmed using alternate extraction and detection (HPLC) methods. Mutations acquired to withstand chloroquine exposure therefore give rise to significant biochemical alterations in the parasite.


Subject(s)
Metabolome , Metabolomics/methods , Plasmodium falciparum/metabolism , Proton Magnetic Resonance Spectroscopy/methods , Animals , Antimalarials/pharmacology , Chloroquine/pharmacology , Chromatography, High Pressure Liquid , Drug Resistance/drug effects , Drug Resistance/genetics , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/parasitology , Female , Host-Parasite Interactions , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation , Oocytes/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Species Specificity , Trophozoites/metabolism , Trophozoites/physiology , Xenopus laevis
20.
ACS Med Chem Lett ; 5(5): 576-81, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24900883

ABSTRACT

The emergence and spread of malaria parasites that are resistant to chloroquine (CQ) has been a disaster for world health. The antihistamine chlorpheniramine (CP) partially resensitizes CQ-resistant (CQR) parasites to CQ but possesses little intrinsic antiplasmodial activity. Mutations in the parasite's CQ resistance transporter (PfCRT) confer resistance to CQ by enabling the protein to transport the drug away from its site of action, and it is thought that resistance-reversers such as CP exert their effect by blocking this CQ transport activity. Here, a series of new structural analogues and homologues of CP have been synthesized. We show that these compounds (along with other in vitro CQ resistance-reversers) inhibit the transport of CQ via a resistance-conferring form of PfCRT expressed in Xenopus laevis oocytes. Furthermore, the level of PfCRT-inhibition was found to correlate well with both the restoration of CQ accumulation and the level of CQ resensitization in CQR parasites.

SELECTION OF CITATIONS
SEARCH DETAIL
...