Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(10): e0274993, 2022.
Article in English | MEDLINE | ID: mdl-36201550

ABSTRACT

The mitochondrion of malaria parasites is an attractive antimalarial drug target, which require mitoribosomes to translate genes encoded in the mitochondrial (mt) DNA. Plasmodium mitoribosomes are composed of highly fragmented ribosomal RNA (rRNA) encoded in the mtDNA. All mitoribosomal proteins (MRPs) and other assembly factors are encoded in the nuclear genome. Here, we have studied one putative assembly factor, RSM22 (Pf3D7_1027200) and one large subunit (LSU) MRP, L23 (Pf3D7_1239100) in Plasmodium falciparum. We show that both proteins localize to the mitochondrion. Conditional knock down (KD) of PfRSM22 or PfMRPL23 leads to reduced cytochrome bc1 complex activity and increased sensitivity to bc1 inhibitors such as atovaquone and ELQ-300. Using RNA sequencing as a tool, we reveal the transcriptomic changes of nuclear and mitochondrial genomes upon KD of these two proteins. In the early phase of KD, while most mt rRNAs and transcripts of putative MRPs were downregulated in the absence of PfRSM22, many mt rRNAs and several MRPs were upregulated after KD of PfMRPL23. The contrast effects in the early phase of KD likely suggests non-redundant roles of PfRSM22 and PfMRPL23 in the assembly of P. falciparum mitoribosomes. At the late time points of KD, loss of PfRSM22 and PfMRPL23 caused defects in many essential metabolic pathways and transcripts related to essential mitochondrial functions, leading to parasite death. In addition, we enlist mitochondrial proteins of unknown function that are likely novel Plasmodium MRPs based on their structural similarity to known MRPs as well as their expression profiles in KD parasites.


Subject(s)
Antimalarials , Malaria, Falciparum , Plasmodium , Antimalarials/therapeutic use , Atovaquone/pharmacology , DNA, Mitochondrial/metabolism , Electron Transport Complex III/metabolism , Humans , Malaria, Falciparum/drug therapy , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Plasmodium/genetics , Plasmodium falciparum , RNA, Ribosomal/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Transcription, Genetic
2.
PLoS One ; 17(8): e0273357, 2022.
Article in English | MEDLINE | ID: mdl-35984838

ABSTRACT

Despite ongoing efforts to control malaria infection, progress in lowering the number of deaths and infections appears to have stalled. The continued high incidence of malaria infection and mortality is in part due to emergence of parasites resistant to frontline antimalarials. This highlights the need for continued identification of novel protein drug targets. Mitochondrial functions in Plasmodium falciparum, the deadliest species of human malaria parasite, are targets of validated antimalarials including atovaquone and proguanil (Malarone). Thus, there has been great interest in identifying other essential mitochondrial proteins as candidates for novel drug targets. Garnering an increased understanding of the proteomic landscape inside the P. falciparum mitochondrion will also allow us to learn about the basic biology housed within this unique organelle. We employed a proximity biotinylation technique and mass spectrometry to identify novel P. falciparum proteins putatively targeted to the mitochondrion. We fused the leader sequence of a mitochondrially targeted chaperone, Hsp60, to the promiscuous biotin ligase TurboID. Through these experiments, we generated a list of 122 "putative mitochondrial" proteins. To verify whether these proteins were indeed mitochondrial, we chose five candidate proteins of interest for localization studies using ectopic expression and tagging of each full-length protein. This allowed us to localize four candidate proteins of unknown function to the mitochondrion, three of which have previously been assessed to be essential. We suggest that phenotypic characterization of these and other proteins from this list of 122 could be fruitful in understanding the basic mitochondrial biology of these parasites and aid antimalarial drug discovery efforts.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria , Antimalarials/therapeutic use , Atovaquone/therapeutic use , Biotinylation , Drug Combinations , Humans , Malaria/parasitology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Proguanil/therapeutic use , Proteomics
3.
Microbiol Spectr ; 10(1): e0015822, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35196803

ABSTRACT

Cholesterol is the most abundant lipid in the erythrocyte. During its blood-stage development, the malaria parasite establishes an active cholesterol gradient across the various membrane systems within the infected erythrocyte. Interestingly, some antimalarial compounds have recently been shown to disrupt cholesterol homeostasis in the intraerythrocytic stages of Plasmodium falciparum. These studies point to the importance of cholesterol for parasite growth. Previously, reduction of cholesterol from the erythrocyte membrane by treatment with methyl-ß-cyclodextrin (MßCD) was shown to inhibit parasite invasion and growth. In addition, MßCD treatment of trophozoite-stage P. falciparum was shown to result in parasite expulsion from the host cell. We have revisited these phenomena by using live video microscopy, ultrastructural analysis, and response to antimalarial compounds. By using time-lapse video microscopy of fluorescently tagged parasites, we show that MßCD treatment for just 30 min causes dramatic expulsion of the trophozoite-stage parasites. This forceful expulsion occurs within 10 s. Remarkably, the plasma membrane of the host cell from which the parasite has been expelled does not appear to be compromised. The parasitophorous vacuolar membrane (PVM) continued to surround the extruded parasite, but the PVM appeared damaged. Treatment with antimalarial compounds targeting PfATP4 or PfNCR1 prevented MßCD-mediated extrusion of the parasites, pointing to a potential role of cholesterol dynamics underlying the expulsion phenomena. We also confirmed the essential role of erythrocyte plasma membrane cholesterol for invasion and growth of P. falciparum. This defect can be partially complemented by cholesterol and desmosterol but not with epicholesterol, revealing stereospecificity underlying cholesterol function. Overall, our studies advance previous observations and reveal unusual cell biological features underlying cholesterol depletion of the infected erythrocyte plasma membrane. IMPORTANCE Malaria remains a major challenge in much of the world. Symptoms of malaria are caused by the growth of parasites belonging to Plasmodium spp. inside the red blood cells (RBCs), leading to their destruction. The parasite depends upon its host for much of its nutritional needs. Cholesterol is a major lipid in the RBC plasma membrane, which is the only source of this lipid for malaria parasites. We have previously shown that certain new antimalarial compounds disrupt cholesterol homeostasis in P. falciparum. Here, we use live time-lapse video microscopy to show dramatic expulsion of the parasite from the host RBC when the cholesterol content of the RBC is reduced. Remarkably, this expulsion is inhibited by the antimalarials that disrupt lipid homeostasis. We also show stereospecificity of cholesterol in supporting parasite growth inside RBC. Overall, these results point to a critical role of cholesterol in the physiology of malaria parasites.


Subject(s)
Cholesterol/metabolism , Erythrocyte Membrane/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Antimalarials/pharmacology , Erythrocyte Membrane/genetics , Erythrocytes/metabolism , Erythrocytes/parasitology , Humans , Malaria, Falciparum/parasitology , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , beta-Cyclodextrins/pharmacology
4.
Article in English | MEDLINE | ID: mdl-33361312

ABSTRACT

The continued emergence of drug-resistant Plasmodium falciparum parasites hinders global attempts to eradicate malaria, emphasizing the need to identify new antimalarial drugs. Attractive targets for chemotherapeutic intervention are the cytochrome (cyt) bc1 complex, which is an essential component of the mitochondrial electron transport chain (mtETC) required for ubiquinone recycling and mitochondrially localized dihydroorotate dehydrogenase (DHODH) critical for de novo pyrimidine synthesis. Despite the essentiality of this complex, resistance to a novel acridone class of compounds targeting cyt bc1 was readily attained, resulting in a parasite strain (SB1-A6) that was panresistant to both mtETC and DHODH inhibitors. Here, we describe the molecular mechanism behind the resistance of the SB1-A6 parasite line, which lacks the common cyt bc1 point mutations characteristic of resistance to mtETC inhibitors. Using Illumina whole-genome sequencing, we have identified both a copy number variation (∼2×) and a single-nucleotide polymorphism (C276F) associated with pfdhodh in SB1-A6. We have characterized the role of both genetic lesions by mimicking the copy number variation via episomal expression of pfdhodh and introducing the identified single nucleotide polymorphism (SNP) using CRISPR-Cas9 and assessed their contributions to drug resistance. Although both of these genetic polymorphisms have been previously identified as contributing to both DSM-1 and atovaquone resistance, SB1-A6 represents a unique genotype in which both alterations are present in a single line, suggesting that the combination contributes to the panresistant phenotype. This novel mechanism of resistance to mtETC inhibition has critical implications for the development of future drugs targeting the bc1 complex or de novo pyrimidine synthesis that could help guide future antimalarial combination therapies and reduce the rapid development of drug resistance in the field.


Subject(s)
Antimalarials , Malaria, Falciparum , Antimalarials/pharmacology , Antimalarials/therapeutic use , DNA Copy Number Variations/genetics , Drug Resistance/genetics , Humans , Malaria, Falciparum/drug therapy , Mitochondria , Plasmodium falciparum/genetics
5.
PLoS One ; 14(4): e0214023, 2019.
Article in English | MEDLINE | ID: mdl-30964863

ABSTRACT

The battle against malaria has been substantially impeded by the recurrence of drug resistance in Plasmodium falciparum, the deadliest human malaria parasite. To counter the problem, novel antimalarial drugs are urgently needed, especially those that target unique pathways of the parasite, since they are less likely to have side effects. The mitochondrial type II NADH dehydrogenase (NDH2) of P. falciparum, PfNDH2 (PF3D7_0915000), has been considered a good prospective antimalarial drug target for over a decade, since malaria parasites lack the conventional multi-subunit NADH dehydrogenase, or Complex I, present in the mammalian mitochondrial electron transport chain (mtETC). Instead, Plasmodium parasites contain a single subunit NDH2, which lacks proton pumping activity and is absent in humans. A significant amount of effort has been expended to develop PfNDH2 specific inhibitors, yet the essentiality of PfNDH2 has not been convincingly verified. Herein, we knocked out PfNDH2 in P. falciparum via a CRISPR/Cas9 mediated approach. Deletion of PfNDH2 does not alter the parasite's susceptibility to multiple mtETC inhibitors, including atovaquone and ELQ-300. We also show that the antimalarial activity of the fungal NDH2 inhibitor HDQ and its new derivative CK-2-68 is due to inhibition of the parasite cytochrome bc1 complex rather than PfNDH2. These compounds directly inhibit the ubiquinol-cytochrome c reductase activity of the malarial bc1 complex. Our results suggest that PfNDH2 is not likely a good antimalarial drug target.


Subject(s)
Antimalarials/pharmacology , Drug Resistance/genetics , NADH Dehydrogenase/genetics , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Animals , Antimalarials/therapeutic use , CRISPR-Cas Systems , Cells, Cultured , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex III/antagonists & inhibitors , Erythrocytes/parasitology , Gene Knockout Techniques , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Mitochondria/drug effects , Mitochondria/enzymology , NADH Dehydrogenase/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Quinolones/pharmacology , Quinolones/therapeutic use
6.
ACS Infect Dis ; 5(4): 550-558, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30638365

ABSTRACT

Lipid homeostasis is essential to the maintenance of life. We previously reported that disruptions of the parasite Na+ homeostasis via inhibition of PfATP4 resulted in elevated cholesterol within the parasite plasma membrane as assessed by saponin sensitivity. A large number of compounds have been shown to target the parasite Na+ homeostasis. We screened 800 compounds from the Malaria and Pathogen Boxes to identify chemotypes that disrupted the parasite plasma membrane lipid homeostasis. Here, we show that the compounds disrupting parasite Na+ homeostasis also induced saponin sensitivity, an indication of parasite lipid homeostasis disruption. Remarkably, 13 compounds were identified that altered the plasma membrane lipid composition independently of the Na+ homeostasis disruption. Further studies suggest that these compounds target the Plasmodium falciparum Niemann-Pick type C1-related (PfNCR1) protein, which is hypothesized to be involved in maintaining plasma membrane lipid composition. PfNCR1, like PfATP4, appears to be targeted by multiple chemotypes with the potential for drug discovery.


Subject(s)
Antimalarials/pharmacology , Cell Membrane/drug effects , Lipid Metabolism/drug effects , Plasmodium falciparum/drug effects , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Erythrocytes/parasitology , Homeostasis/drug effects , Humans , Lipids/chemistry , Malaria, Falciparum/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Sodium/metabolism
7.
J Biol Chem ; 293(21): 8128-8137, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29626096

ABSTRACT

The phylum Apicomplexa contains a group of protozoa causing diseases in humans and livestock. Plasmodium spp., the causative agent of malaria, contains a mitochondrion that is very divergent from that of their hosts. The malarial mitochondrion is a clinically validated target for the antimalarial drug atovaquone, which specifically blocks the electron transfer activity of the bc1 complex of the mitochondrial electron transport chain (mtETC). Most mtETC proteins are nuclear-encoded and imported from the cytosol, but three key protein subunits are encoded in the Plasmodium mitochondrial genome: cyt b, COXI, and COXIII. They are translated inside the mitochondrion by mitochondrial ribosomes (mitoribosomes). Here, we characterize the function of one large mitoribosomal protein in Plasmodium falciparum, PfmRPL13. We found that PfmRPL13 localizes to the parasite mitochondrion and is refractory to genetic knockout. Ablation of PfmRPL13 using a conditional knockdown system (TetR-DOZI-aptamer) caused a series of adverse events in the parasite, including mtETC deficiency, loss of mitochondrial membrane potential (Δψm), and death. The PfmRPL13 knockdown parasite also became hypersensitive to proguanil, a drug proposed to target an alternative process for maintaining Δψm Surprisingly, transmission EM revealed that PfmRPL13 disruption also resulted in an unusually elongated and branched mitochondrion. The growth arrest of the knockdown parasite could be rescued with a second copy of PfmRPL13, but not by supplementation with decylubiquinone or addition of a yeast dihydroorotate dehydrogenase gene. In summary, we provide first and direct evidence that mitoribosomes are essential for malaria parasites to maintain the structural and functional integrity of the mitochondrion.


Subject(s)
Antimalarials/pharmacology , Malaria/drug therapy , Mitochondria/chemistry , Mitochondria/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Plasmodium falciparum/metabolism , Ribosomal Proteins/metabolism , Electron Transport , Genome, Mitochondrial , Humans , Malaria/metabolism , Malaria/parasitology , Membrane Potential, Mitochondrial , Mitochondria/drug effects , Oxidoreductases Acting on CH-CH Group Donors/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/isolation & purification , Ribosomal Proteins/genetics
8.
Article in English | MEDLINE | ID: mdl-27799215

ABSTRACT

Caged Garcinia xanthones (CGXs) constitute a family of natural products that are produced by tropical/subtropical trees of the genus Garcinia CGXs have a unique chemical architecture, defined by the presence of a caged scaffold at the C ring of a xanthone moiety, and exhibit a broad range of biological activities. Here we show that synthetic CGXs exhibit antimalarial activity against Plasmodium falciparum, the causative parasite of human malaria, at the intraerythrocytic stages. Their activity can be substantially improved by attaching a triphenylphosphonium group at the A ring of the caged xanthone. Specifically, CR135 and CR142 were found to be highly effective antimalarial inhibitors, with 50% effective concentrations as low as ∼10 nM. CGXs affect malaria parasites at multiple intraerythrocytic stages, with mature stages (trophozoites and schizonts) being more vulnerable than immature rings. Within hours of CGX treatment, malaria parasites display distinct morphological changes, significant reduction of parasitemia (the percentage of infected red blood cells), and aberrant mitochondrial fragmentation. CGXs do not, however, target the mitochondrial electron transport chain, the target of the drug atovaquone and several preclinical candidates. CGXs are cytotoxic to human HEK293 cells at the low micromolar level, which results in a therapeutic window of around 150-fold for the lead compounds. In summary, we show that CGXs are potent antimalarial compounds with structures distinct from those of previously reported antimalarial inhibitors. Our results highlight the potential to further develop Garcinia natural product derivatives as novel antimalarial agents.


Subject(s)
Antimalarials/pharmacology , Garcinia/chemistry , Xanthones/pharmacology , Antimalarials/chemistry , Antimalarials/therapeutic use , HEK293 Cells , Humans , Mitochondria/drug effects , Molecular Structure , Parasitemia/drug therapy , Parasitemia/parasitology , Plasmodium falciparum/drug effects , Schizonts/drug effects , Structure-Activity Relationship , Trophozoites/drug effects , Xanthones/chemistry , Xanthones/therapeutic use
9.
Antimicrob Agents Chemother ; 60(8): 4853-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27270285

ABSTRACT

Antimalarial combination therapies play a crucial role in preventing the emergence of drug-resistant Plasmodium parasites. Although artemisinin-based combination therapies (ACTs) comprise the majority of these formulations, inhibitors of the mitochondrial cytochrome bc1 complex (cyt bc1) are among the few compounds that are effective for both acute antimalarial treatment and prophylaxis. There are two known sites for inhibition within cyt bc1: atovaquone (ATV) blocks the quinol oxidase (Qo) site of cyt bc1, while some members of the endochin-like quinolone (ELQ) family, including preclinical candidate ELQ-300, inhibit the quinone reductase (Qi) site and retain full potency against ATV-resistant Plasmodium falciparum strains with Qo site mutations. Here, we provide the first in vivo comparison of ATV, ELQ-300, and combination therapy consisting of ATV plus ELQ-300 (ATV:ELQ-300), using P. yoelii murine models of malaria. In our monotherapy assessments, we found that ATV functioned as a single-dose curative compound in suppressive tests whereas ELQ-300 demonstrated a unique cumulative dosing effect that successfully blocked recrudescence even in a high-parasitemia acute infection model. ATV:ELQ-300 therapy was highly synergistic, and the combination was curative with a single combined dose of 1 mg/kg of body weight. Compared to the ATV:proguanil (Malarone) formulation, ATV:ELQ-300 was more efficacious in multiday, acute infection models and was equally effective at blocking the emergence of ATV-resistant parasites. Ultimately, our data suggest that dual-site inhibition of cyt bc1 is a valuable strategy for antimalarial combination therapy and that Qi site inhibitors such as ELQ-300 represent valuable partner drugs for the clinically successful Qo site inhibitor ATV.


Subject(s)
Antimalarials/pharmacology , Atovaquone/pharmacology , Electron Transport Complex III/antagonists & inhibitors , Malaria, Falciparum/drug therapy , Quinolones/pharmacology , Animals , Drug Combinations , Drug Therapy, Combination/methods , Female , Mice , Parasitemia/drug therapy , Plasmodium falciparum/drug effects , Proguanil/pharmacology
10.
PLoS Pathog ; 12(5): e1005647, 2016 05.
Article in English | MEDLINE | ID: mdl-27227970

ABSTRACT

Among the several new antimalarials discovered over the past decade are at least three clinical candidate drugs, each with a distinct chemical structure, that disrupt Na+ homeostasis resulting in a rapid increase in intracellular Na+ concentration ([Na+]i) within the erythrocytic stages of Plasmodium falciparum. At present, events triggered by Na+ influx that result in parasite demise are not well-understood. Here we report effects of two such drugs, a pyrazoleamide and a spiroindolone, on intraerythrocytic P. falciparum. Within minutes following the exposure to these drugs, the trophozoite stage parasite, which normally contains little cholesterol, was made permeant by cholesterol-dependent detergents, suggesting it acquired a substantial amount of the lipid. Consistently, the merozoite surface protein 1 and 2 (MSP1 and MSP2), glycosylphosphotidylinositol (GPI)-anchored proteins normally uniformly distributed in the parasite plasma membrane, coalesced into clusters. These alterations were not observed following drug treatment of P. falciparum parasites adapted to grow in a low [Na+] growth medium. Both cholesterol acquisition and MSP1 coalescence were reversible upon the removal of the drugs, implicating an active process of cholesterol exclusion from trophozoites that we hypothesize is inhibited by high [Na+]i. Electron microscopy of drug-treated trophozoites revealed substantial morphological changes normally seen at the later schizont stage including the appearance of partial inner membrane complexes, dense organelles that resemble "rhoptries" and apparent nuclear division. Together these results suggest that [Na+]i disruptor drugs by altering levels of cholesterol in the parasite, dysregulate trophozoite to schizont development and cause parasite demise.


Subject(s)
Antimalarials/pharmacology , Cholesterol/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Plasmodium falciparum/drug effects , Sodium/metabolism , Blotting, Western , Flow Cytometry , Fluorescent Antibody Technique , Humans , Microscopy, Electron, Transmission , Plasmodium falciparum/metabolism
11.
PLoS One ; 11(3): e0152197, 2016.
Article in English | MEDLINE | ID: mdl-27015086

ABSTRACT

Coenzyme Q (CoQ, ubiquinone) is a central electron carrier in mitochondrial respiration. CoQ is synthesized through multiple steps involving a number of different enzymes. The prevailing view that the CoQ used in respiration exists as a free pool that diffuses throughout the mitochondrial inner membrane bilayer has recently been challenged. In the yeast Saccharomyces cerevisiae, deletion of the gene encoding Coq10p results in respiration deficiency without inhibiting the synthesis of CoQ, suggesting that the Coq10 protein is critical for the delivery of CoQ to the site(s) of respiration. The precise mechanism by which this is achieved remains unknown at present. We have identified a Plasmodium orthologue of Coq10 (PfCoq10), which is predominantly expressed in trophozoite-stage parasites, and localizes to the parasite mitochondrion. Expression of PfCoq10 in the S. cerevisiae coq10 deletion strain restored the capability of the yeast to grow on respiratory substrates, suggesting a remarkable functional conservation of this protein over a vast evolutionary distance, and despite a relatively low level of amino acid sequence identity. As the antimalarial drug atovaquone acts as a competitive inhibitor of CoQ, we assessed whether over-expression of PfCoq10 altered the atovaquone sensitivity in parasites and in yeast mitochondria, but found no alteration of its activity.


Subject(s)
Carrier Proteins/genetics , Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Ubiquinone/analogs & derivatives , Atovaquone/administration & dosage , Carrier Proteins/biosynthesis , Gene Expression Regulation/drug effects , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Mitochondria/drug effects , Mitochondria/genetics , Plasmodium falciparum/pathogenicity , Respiration/drug effects , Respiration/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Ubiquinone/biosynthesis , Ubiquinone/deficiency , Ubiquinone/genetics
12.
Cell Rep ; 11(1): 164-74, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25843709

ABSTRACT

New antimalarial drugs are urgently needed to control drug-resistant forms of the malaria parasite Plasmodium falciparum. Mitochondrial electron transport is the target of both existing and new antimalarials. Herein, we describe 11 genetic knockout (KO) lines that delete six of the eight mitochondrial tricarboxylic acid (TCA) cycle enzymes. Although all TCA KOs grew normally in asexual blood stages, these metabolic deficiencies halted life-cycle progression in later stages. Specifically, aconitase KO parasites arrested as late gametocytes, whereas α-ketoglutarate-dehydrogenase-deficient parasites failed to develop oocysts in the mosquitoes. Mass spectrometry analysis of (13)C-isotope-labeled TCA mutant parasites showed that P. falciparum has significant flexibility in TCA metabolism. This flexibility manifested itself through changes in pathway fluxes and through altered exchange of substrates between cytosolic and mitochondrial pools. Our findings suggest that mitochondrial metabolic plasticity is essential for parasite development.


Subject(s)
Enzymes/genetics , Malaria, Falciparum/genetics , Mitochondria/metabolism , Plasmodium falciparum/genetics , Tricarboxylic Acids/metabolism , Animals , Antimalarials/chemistry , Antimalarials/isolation & purification , Antimalarials/metabolism , Citric Acid Cycle/genetics , Enzymes/metabolism , Erythrocytes/metabolism , Gene Knockout Techniques , Humans , Life Cycle Stages , Malaria, Falciparum/drug therapy , Malaria, Falciparum/enzymology , Malaria, Falciparum/parasitology , Mitochondria/pathology , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Plasmodium falciparum/pathogenicity
13.
Am J Trop Med Hyg ; 92(6): 1195-201, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25918204

ABSTRACT

Single-dose therapies for malaria have been proposed as a way to reduce the cost and increase the effectiveness of antimalarial treatment. However, no compound to date has shown single-dose activity against both the blood-stage Plasmodium parasites that cause disease and the liver-stage parasites that initiate malaria infection. Here, we describe a subset of cytochrome bc1 (cyt bc1) inhibitors, including the novel 4(1H)-quinolone ELQ-400, with single-dose activity against liver, blood, and transmission-stage parasites in mouse models of malaria. Although cyt bc1 inhibitors are generally classified as slow-onset antimalarials, we found that a single dose of ELQ-400 rapidly induced stasis in blood-stage parasites, which was associated with a rapid reduction in parasitemia in vivo. ELQ-400 also exhibited a low propensity for drug resistance and was active against atovaquone-resistant P. falciparum strains with point mutations in cyt bc1. Ultimately, ELQ-400 shows that cyt bc1 inhibitors can function as single-dose, blood-stage antimalarials and is the first compound to provide combined treatment, prophylaxis, and transmission blocking activity for malaria after a single oral administration. This remarkable multi-stage efficacy suggests that metabolic therapies, including cyt bc1 inhibitors, may be valuable additions to the collection of single-dose antimalarials in current development.


Subject(s)
Antimalarials/therapeutic use , Electron Transport Complex III/antagonists & inhibitors , Malaria, Falciparum/drug therapy , Phenyl Ethers/therapeutic use , Quinolones/therapeutic use , Animals , Antimalarials/administration & dosage , Drug Resistance , Electron Transport Complex III/metabolism , Female , Mice , Parasitemia/drug therapy , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects
14.
Antimicrob Agents Chemother ; 59(4): 1977-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605352

ABSTRACT

The cytochrome bc1 complex (cyt bc1) is the third component of the mitochondrial electron transport chain and is the target of several potent antimalarial compounds, including the naphthoquinone atovaquone (ATV) and the 4(1H)-quinolone ELQ-300. Mechanistically, cyt bc1 facilitates the transfer of electrons from ubiquinol to cytochrome c and contains both oxidative (Qo) and reductive (Qi) catalytic sites that are amenable to small-molecule inhibition. Although many antimalarial compounds, including ATV, effectively target the Qo site, it has been challenging to design selective Qi site inhibitors with the ability to circumvent clinical ATV resistance, and little is known about how chemical structure contributes to site selectivity within cyt bc1. Here, we used the proposed Qi site inhibitor ELQ-300 to generate a drug-resistant Plasmodium falciparum clone containing an I22L mutation at the Qi region of cyt b. Using this D1 clone and the Y268S Qo mutant strain, P. falciparum Tm90-C2B, we created a structure-activity map of Qi versus Qo site selectivity for a series of endochin-like 4(1H)-quinolones (ELQs). We found that Qi site inhibition was associated with compounds containing 6-position halogens or aryl 3-position side chains, while Qo site inhibition was favored by 5,7-dihalogen groups or 7-position substituents. In addition to identifying ELQ-300 as a preferential Qi site inhibitor, our data suggest that the 4(1H)-quinolone scaffold is compatible with binding to either site of cyt bc1 and that minor chemical changes can influence Qo or Qi site inhibition by the ELQs.


Subject(s)
Antimalarials/pharmacology , Electron Transport Complex III/antagonists & inhibitors , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Quinolones/pharmacology , Animals , Cytochromes b/genetics , Cytochromes b/metabolism , Drug Resistance , Electron Transport Complex III/genetics , Models, Molecular , Mutation/genetics , Plasmodium falciparum/genetics , Protein Binding , Structure-Activity Relationship
15.
Nat Commun ; 5: 5521, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25422853

ABSTRACT

The quest for new antimalarial drugs, especially those with novel modes of action, is essential in the face of emerging drug-resistant parasites. Here we describe a new chemical class of molecules, pyrazoleamides, with potent activity against human malaria parasites and showing remarkably rapid parasite clearance in an in vivo model. Investigations involving pyrazoleamide-resistant parasites, whole-genome sequencing and gene transfers reveal that mutations in two proteins, a calcium-dependent protein kinase (PfCDPK5) and a P-type cation-ATPase (PfATP4), are necessary to impart full resistance to these compounds. A pyrazoleamide compound causes a rapid disruption of Na(+) regulation in blood-stage Plasmodium falciparum parasites. Similar effect on Na(+) homeostasis was recently reported for spiroindolones, which are antimalarials of a chemical class quite distinct from pyrazoleamides. Our results reveal that disruption of Na(+) homeostasis in malaria parasites is a promising mode of antimalarial action mediated by at least two distinct chemical classes.


Subject(s)
Amides/pharmacology , Antimalarials/pharmacology , Benzimidazoles/pharmacology , Erythrocytes/parasitology , Malaria/parasitology , Plasmodium falciparum/drug effects , Pyrazoles/pharmacology , Sodium/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Female , Homeostasis/drug effects , Humans , Male , Plasmodium berghei/drug effects , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Protozoan Proteins
16.
J Biol Chem ; 289(50): 34827-37, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25352601

ABSTRACT

Heme is an essential cofactor for aerobic organisms. Its redox chemistry is central to a variety of biological functions mediated by hemoproteins. In blood stages, malaria parasites consume most of the hemoglobin inside the infected erythrocytes, forming nontoxic hemozoin crystals from large quantities of heme released during digestion. At the same time, the parasites possess a heme de novo biosynthetic pathway. This pathway in the human malaria parasite Plasmodium falciparum has been considered essential and is proposed as a potential drug target. However, we successfully disrupted the first and last genes of the pathway, individually and in combination. These knock-out parasite lines, lacking 5-aminolevulinic acid synthase and/or ferrochelatase (FC), grew normally in blood-stage culture and exhibited no changes in sensitivity to heme-related antimalarial drugs. We developed a sensitive LC-MS/MS assay to monitor stable isotope incorporation into heme from its precursor 5-[(13)C4]aminolevulinic acid, and this assay confirmed that de novo heme synthesis was ablated in FC knock-out parasites. Disrupting the FC gene also caused no defects in gametocyte generation or maturation but resulted in a greater than 70% reduction in male gamete formation and completely prevented oocyst formation in female Anopheles stephensi mosquitoes. Our data demonstrate that the heme biosynthesis pathway is not essential for asexual blood-stage growth of P. falciparum parasites but is required for mosquito transmission. Drug inhibition of pathway activity is therefore unlikely to provide successful antimalarial therapy. These data also suggest the existence of a parasite mechanism for scavenging host heme to meet metabolic needs.


Subject(s)
Anopheles/parasitology , Erythrocytes/parasitology , Heme/biosynthesis , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , 5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/genetics , Animals , Female , Ferrochelatase/genetics , Gene Knockout Techniques , Heme/metabolism , Humans , Male , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Tandem Mass Spectrometry
18.
Sci Transl Med ; 5(177): 177ra37, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23515079

ABSTRACT

The goal for developing new antimalarial drugs is to find a molecule that can target multiple stages of the parasite's life cycle, thus impacting prevention, treatment, and transmission of the disease. The 4(1H)-quinolone-3-diarylethers are selective potent inhibitors of the parasite's mitochondrial cytochrome bc1 complex. These compounds are highly active against the human malaria parasites Plasmodium falciparum and Plasmodium vivax. They target both the liver and blood stages of the parasite as well as the forms that are crucial for disease transmission, that is, the gametocytes, the zygote, the ookinete, and the oocyst. Selected as a preclinical candidate, ELQ-300 has good oral bioavailability at efficacious doses in mice, is metabolically stable, and is highly active in blocking transmission in rodent models of malaria. Given its predicted low dose in patients and its predicted long half-life, ELQ-300 has potential as a new drug for the treatment, prevention, and, ultimately, eradication of human malaria.


Subject(s)
Antimalarials/pharmacology , Quinolones/pharmacology , Animals , Antimalarials/chemistry , Atovaquone/chemistry , Atovaquone/pharmacology , Drug Resistance , Drug Synergism , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Mice , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects , Proguanil/chemistry , Proguanil/pharmacology , Pyridones/chemistry , Pyridones/pharmacology , Quinolones/chemistry
19.
Mol Biochem Parasitol ; 185(1): 48-51, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22640832

ABSTRACT

We have shown that transgenic Plasmodium falciparum parasites expressing the yeast DHODH (dihydroorotate dehydrogenase) are independent of the mtETC (mitochondrial electron transport chain), suggesting that they might not need the mitochondrial genome (mtDNA), since it only encodes three protein subunits belonging to the mtETC and fragmentary ribosomal RNA molecules. Disrupting the mitochondrial RNA polymerase (mtRNAP), which is critical for mtDNA replication and transcription, might then cause the generation of a ρ(0) parasite line lacking mtDNA. We made multiple attempts to disrupt the mtRNAP gene by double crossover recombination methods in parasite lines expressing yDHODH either episomally or integrated in the genome, but were unable to produce the desired knockout. We verified that the mtRNAP gene was accessible to recombination by successfully integrating a triple HA tag at the 3' end via single cross-over recombination. These studies suggest that mtRNAP is essential even in mtETC-independent P. falciparum parasites.


Subject(s)
DNA, Protozoan/genetics , DNA-Directed RNA Polymerases/metabolism , Erythrocytes/parasitology , Genes, Mitochondrial , Mitochondria/enzymology , Plasmodium falciparum/enzymology , Animals , Crossing Over, Genetic , DNA, Mitochondrial/genetics , Dihydroorotate Dehydrogenase , Electron Transport , Gene Knockout Techniques/methods , Genes, Protozoan , Life Cycle Stages , Mitochondrial Proteins/metabolism , Organisms, Genetically Modified/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Plasmids/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Transfection
20.
J Biol Chem ; 286(48): 41312-41322, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21984828

ABSTRACT

The rotary nanomotor ATP synthase is a central player in the bioenergetics of most organisms. Yet the role of ATP synthase in malaria parasites has remained unclear, as blood stages of Plasmodium falciparum appear to derive ATP largely through glycolysis. Also, genes for essential subunits of the F(O) sector of the complex could not be detected in the parasite genomes. Here, we have used molecular genetic and immunological tools to investigate the localization, complex formation, and functional significance of predicted ATP synthase subunits in P. falciparum. We generated transgenic P. falciparum lines expressing seven epitope-tagged canonical ATP synthase subunits, revealing localization of all but one of the subunits to the mitochondrion. Blue native gel electrophoresis of P. falciparum mitochondrial membranes suggested the molecular mass of the ATP synthase complex to be greater than 1 million daltons. This size is consistent with the complex being assembled as a dimer in a manner similar to the complexes observed in other eukaryotic organisms. This observation also suggests the presence of previously unknown subunits in addition to the canonical subunits in P. falciparum ATP synthase complex. Our attempts to disrupt genes encoding ß and γ subunits were unsuccessful, suggesting an essential role played by the ATP synthase complex in blood stages of P. falciparum. These studies suggest that, despite some unconventional features and its minimal contribution to ATP synthesis, P. falciparum ATP synthase is localized to the parasite mitochondrion, assembled as a large dimeric complex, and is likely essential for parasite survival.


Subject(s)
Merozoites/enzymology , Mitochondria/enzymology , Multienzyme Complexes/metabolism , Plasmodium falciparum/enzymology , Proton-Translocating ATPases/metabolism , Protozoan Proteins/metabolism , Glycolysis/physiology , Mitochondria/genetics , Multienzyme Complexes/genetics , Plasmodium falciparum/genetics , Proton-Translocating ATPases/genetics , Protozoan Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...