Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
bioRxiv ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38187525

ABSTRACT

Artemisinin (ART) combination therapies have been critical in reducing malaria morbidity and mortality, but these important drugs are threatened by growing resistance associated with mutations in Pfcoronin and Pfkelch13 . Here, we describe the mechanism of Pfcoronin -mediated ART resistance. Pf Coronin interacts with Pf Actin and localizes to the parasite plasma membrane (PPM), the digestive vacuole (DV) membrane, and membrane of a newly identified preDV compartment-all structures involved in the trafficking of hemoglobin from the RBC for degradation in the DV. Pfcoronin mutations alter Pf Actin homeostasis and impair the development and morphology of the preDV. Ultimately, these changes are associated with decreased uptake of red blood cell cytosolic contents by ring-stage Plasmodium falciparum . Previous work has identified decreased hemoglobin uptake as the mechanism of Pfkelch 13-mediated ART resistance. This work demonstrates that Pf Coronin appears to act via a parallel pathway. For both Pfkelch13 -mediated and Pfcoronin -mediated ART resistance, we hypothesize that the decreased hemoglobin uptake in ring stage parasites results in less heme-based activation of the artemisinin endoperoxide ring and reduced cytocidal activity. This study deepens our understanding of ART resistance, as well as hemoglobin uptake and development of the DV in early-stage parasites.

2.
Nat Commun ; 14(1): 1455, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36927839

ABSTRACT

Identifying how small molecules act to kill malaria parasites can lead to new "chemically validated" targets. By pressuring Plasmodium falciparum asexual blood stage parasites with three novel structurally-unrelated antimalarial compounds (MMV665924, MMV019719 and MMV897615), and performing whole-genome sequence analysis on resistant parasite lines, we identify multiple mutations in the P. falciparum acyl-CoA synthetase (ACS) genes PfACS10 (PF3D7_0525100, M300I, A268D/V, F427L) and PfACS11 (PF3D7_1238800, F387V, D648Y, and E668K). Allelic replacement and thermal proteome profiling validates PfACS10 as a target of these compounds. We demonstrate that this protein is essential for parasite growth by conditional knockdown and observe increased compound susceptibility upon reduced expression. Inhibition of PfACS10 leads to a reduction in triacylglycerols and a buildup of its lipid precursors, providing key insights into its function. Analysis of the PfACS11 gene and its mutations point to a role in mediating resistance via decreased protein stability.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Plasmodium falciparum/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Antimalarials/pharmacology , Antimalarials/therapeutic use , Mutation , Ligases/metabolism
3.
PLoS Pathog ; 18(6): e1010609, 2022 06.
Article in English | MEDLINE | ID: mdl-35687594

ABSTRACT

The spread of insecticide resistance in Anopheles mosquitoes and drug resistance in Plasmodium parasites is contributing to a global resurgence of malaria, making the generation of control tools that can overcome these roadblocks an urgent public health priority. We recently showed that the transmission of Plasmodium falciparum parasites can be efficiently blocked when exposing Anopheles gambiae females to antimalarials deposited on a treated surface, with no negative consequences on major components of mosquito fitness. Here, we demonstrate this approach can overcome the hurdles of insecticide resistance in mosquitoes and drug resistant in parasites. We show that the transmission-blocking efficacy of mosquito-targeted antimalarials is maintained when field-derived, insecticide resistant Anopheles are exposed to the potent cytochrome b inhibitor atovaquone, demonstrating that this drug escapes insecticide resistance mechanisms that could potentially interfere with its function. Moreover, this approach prevents transmission of field-derived, artemisinin resistant P. falciparum parasites (Kelch13 C580Y mutant), proving that this strategy could be used to prevent the spread of parasite mutations that induce resistance to front-line antimalarials. Atovaquone is also highly effective at limiting parasite development when ingested by mosquitoes in sugar solutions, including in ongoing infections. These data support the use of mosquito-targeted antimalarials as a promising tool to complement and extend the efficacy of current malaria control interventions.


Subject(s)
Anopheles , Antimalarials , Malaria, Falciparum , Malaria , Plasmodium , Animals , Anopheles/parasitology , Antimalarials/pharmacology , Atovaquone/pharmacology , Female , Malaria/parasitology , Malaria/prevention & control , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/genetics
4.
J Infect Dis ; 223(10): 1822-1830, 2021 05 28.
Article in English | MEDLINE | ID: mdl-31875909

ABSTRACT

BACKGROUND: Plasmodium falciparum transmission depends on mature gametocytes that can be ingested by mosquitoes taking a blood meal on human skin. Although gametocyte skin sequestration has long been hypothesized as important contributor to efficient malaria transmission, this has never been formally tested. METHODS: In naturally infected gametocyte carriers from Burkina Faso, we assessed infectivity to mosquitoes by direct skin feeding and membrane feeding. We directly quantified male and female gametocytes and asexual parasites in finger-prick and venous blood samples, skin biopsy samples, and in of mosquitoes that fed on venous blood or directly on skin. Gametocytes were visualized in skin tissue with confocal microscopy. RESULTS: Although more mosquitoes became infected when feeding directly on skin then when feeding on venous blood (odds ratio, 2.01; 95% confidence interval, 1.21-3.33; P = .007), concentrations of gametocytes were not higher in the subdermal skin vasculature than in other blood compartments; only sparse gametocytes were observed in skin tissue. DISCUSSION: Our data strongly suggest that there is no significant skin sequestration of P. falciparum gametocytes. Gametocyte densities in peripheral blood are thus informative for predicting onward transmission potential to mosquitoes and can be used to target and monitor malaria elimination initiatives.


Subject(s)
Anopheles , Malaria, Falciparum , Animals , Anopheles/parasitology , Burkina Faso , Humans , Malaria, Falciparum/epidemiology , Plasmodium falciparum
5.
PLoS Genet ; 16(12): e1009266, 2020 12.
Article in English | MEDLINE | ID: mdl-33370279

ABSTRACT

Malaria continues to impose a significant health burden in the continent of Africa with 213 million cases in 2018 alone, representing 93% of cases worldwide. Because of high transmission of malaria within the continent, the selection pressures to develop drug resistance in African parasites are distinct compared to the rest of the world. In light of the spread of resistance to artemisinin conferred by the C580Y mutation in the PfKelch13 propeller domain in Southeast Asia, and its independent emergence in South America, it is important to study genetic determinants of resistance in the African context using African parasites. Through in vitro evolution of Senegalese parasites, we had previously generated the artemisinin-resistant parasites Pikine_R and Thiès_R and established pfcoronin mutations to be sufficient to confer artemisinin resistance in the standard ring-stage survival assay (RSA). In the current study, we used genetic analysis of revertants to demonstrate pfcoronin to be the major driver of elevated RSA in the artemisinin-resistant parasites Pikine_R and Thiès_R evolved in vitro. We interrogated the role of a second gene PF3D7_1433800, which also had mutations in both the Pikine_R and Thiès_R selected lines, but found no evidence of a contribution to reduced susceptibility in the RSA survival assay. Nevertheless, our genetic analysis demonstrates that parasite genetic background is important in the level of pfcoronin mediated RSA survival, and therefore we cannot rule out a role for PF3D7_1433800 in other genetic backgrounds. Finally, we tested the potential synergy between the mutations of pfcoronin and pfkelch13 through the generation of single and double mutants in the Pikine genetic background and found that the contribution of pfcoronin to reduced susceptibility is masked by the presence of pfkelch13. This phenomenon was also observed in the 3D7 background, suggesting that pfcoronin may mediate its effects via the same pathway as pfkelch13. Investigating the biology of proteins containing the beta-propeller domain could further elucidate the different pathways that the parasite could use to attain resistance.


Subject(s)
Drug Resistance , Genetic Background , Microfilament Proteins/genetics , Mutation , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Antimalarials/pharmacology , Artemisinins/pharmacology , Kelch Repeat , Microfilament Proteins/chemistry , Plasmodium falciparum/drug effects , Protozoan Proteins/chemistry
6.
J Biomed Opt ; 25(1): 1-11, 2019 12.
Article in English | MEDLINE | ID: mdl-31849205

ABSTRACT

Hemozoin, the heme detoxification end product in malaria parasites during their growth in the red blood cells (RBCs), serves as an important marker for diagnosis and treatment target of malaria disease. However, the current method for hemozoin-targeted drug screening mainly relies on in vitro ß-hematin inhibition assays, which may lead to false-positive events due to under-representation of the real hemozoin crystal. Quantitative in situ imaging of hemozoin is highly desired for high-throughput screening of antimalarial drugs and for elucidating the mechanisms of antimalarial drugs. We present transient absorption (TA) imaging as a high-speed single-cell analysis platform with chemical selectivity to hemozoin. We first demonstrated that TA microscopy is able to identify ß-hematin, the artificial form of hemozoin, from the RBCs. We further utilized time-resolved TA imaging to in situ discern hemozoin from malaria-infected RBCs with optimized imaging conditions. Finally, we quantitatively analyzed the hemozoin amount in RBCs at different infection stages by single-shot TA imaging. These results highlight the potential of TA imaging for efficient antimalarial drug screening and drug mechanism investigation.


Subject(s)
Erythrocytes/metabolism , Hemeproteins/metabolism , Microscopy/methods , Animals , Antimalarials/pharmacology , Crystallization , Drug Evaluation, Preclinical , Erythrocytes/parasitology , Hemeproteins/analysis , Hemeproteins/chemistry , High-Throughput Screening Assays , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Microscopy, Electron, Scanning , Optical Phenomena , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Single-Cell Analysis/methods
7.
ACS Infect Dis ; 5(4): 515-520, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30773881

ABSTRACT

We have previously identified the cytoplasmic prolyl tRNA synthetase in Plasmodium falciparum as the functional target of the natural product febrifugine and its synthetic analogue halofuginone (HFG), one of the most potent antimalarials discovered to date. However, our studies also discovered that short-term treatment of asexual blood stage P. falciparum with HFG analogues causes a 20-fold increase in intracellular proline, termed the adaptive proline response (APR), which renders parasites tolerant to HFG. This novel resistance phenotype lacks an apparent genetic basis but remains stable after drug withdrawal. On the basis of our findings that HFG treatment induces eIF2α phosphorylation, a sensitive marker and mediator of cellular stress, we here investigate if eIF2α-signaling is functionally linked to the APR. In our comparative studies using a parasite line lacking PfeIK1, the Plasmodium orthologue of the eIF2α-kinase GCN2 that mediates amino acid deprivation sensing, we show that HFG activity and the APR are independent from PfeIK1 and eIF2α signaling.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Plasmodium falciparum/metabolism , Proline/metabolism , Protozoan Proteins/metabolism , Amino Acyl-tRNA Synthetases/genetics , Antimalarials/pharmacology , Drug Resistance , Eukaryotic Initiation Factor-2/genetics , Humans , Malaria, Falciparum/parasitology , Phosphorylation , Piperidines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Quinazolinones/pharmacology , Signal Transduction/drug effects
8.
Proc Natl Acad Sci U S A ; 115(50): 12799-12804, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30420498

ABSTRACT

Drug resistance is an obstacle to global malaria control, as evidenced by the recent emergence and rapid spread of delayed artemisinin (ART) clearance by mutant forms of the PfKelch13 protein in Southeast Asia. Identifying genetic determinants of ART resistance in African-derived parasites is important for surveillance and for understanding the mechanism of resistance. In this study, we carried out long-term in vitro selection of two recently isolated West African parasites (from Pikine and Thiès, Senegal) with increasing concentrations of dihydroartemisinin (DHA), the biologically active form of ART, over a 4-y period. We isolated two parasite clones, one from each original isolate, that exhibited enhanced survival to DHA in the ring-stage survival assay. Whole-genome sequence analysis identified 10 mutations in seven different genes. We chose to focus on the gene encoding PfCoronin, a member of the WD40-propeller domain protein family, because mutations in this gene occurred in both independent selections, and the protein shares the ß-propeller motif with PfKelch13 protein. For functional validation, when pfcoronin mutations were introduced into the parental parasites by CRISPR/Cas9-mediated gene editing, these mutations were sufficient to reduce ART susceptibility in the parental lines. The discovery of a second gene for ART resistance may yield insights into the molecular mechanisms of resistance. It also suggests that pfcoronin mutants could emerge as a nonkelch13 type of resistance to ART in natural settings.


Subject(s)
4-Butyrolactone/analogs & derivatives , Artemisinins/pharmacology , Microfilament Proteins/genetics , Mutation/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , 4-Butyrolactone/genetics , Antimalarials/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Drug Resistance/genetics , Gene Editing/methods , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , WD40 Repeats/genetics
9.
Nat Commun ; 9(1): 1769, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720620

ABSTRACT

Multidrug resistant Plasmodium falciparum in Southeast Asia endangers regional malaria elimination and threatens to spread to other malaria endemic areas. Understanding mechanisms of piperaquine (PPQ) resistance is crucial for tracking its emergence and spread, and to develop effective strategies for overcoming it. Here we analyze a mechanism of PPQ resistance in Cambodian parasites. Isolates exhibit a bimodal dose-response curve when exposed to PPQ, with the area under the curve quantifying their survival in vitro. Increased copy number for plasmepsin II and plasmepsin III appears to explain enhanced survival when exposed to PPQ in most, but not all cases. A panel of isogenic subclones reinforces the importance of plasmepsin II-III copy number to enhanced PPQ survival. We conjecture that factors producing increased parasite survival under PPQ exposure in vitro may drive clinical PPQ failures in the field.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Drug Resistance/genetics , Gene Dosage , Plasmodium falciparum/drug effects , Protozoan Proteins/genetics , Quinolines/pharmacology , Antimalarials/pharmacology , Aspartic Acid Endopeptidases/metabolism , Cambodia , Cell Survival/drug effects , Cell Survival/genetics , DNA Copy Number Variations , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Plasmodium falciparum/cytology , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Whole Genome Sequencing
10.
Mol Biol Evol ; 35(7): 1678-1689, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29722884

ABSTRACT

Detecting de novo mutations in viral and bacterial pathogens enables researchers to reconstruct detailed networks of disease transmission and is a key technique in genomic epidemiology. However, these techniques have not yet been applied to the malaria parasite, Plasmodium falciparum, in which a larger genome, slower generation times, and a complex life cycle make them difficult to implement. Here, we demonstrate the viability of de novo mutation studies in P. falciparum for the first time. Using a combination of sequencing, library preparation, and genotyping methods that have been optimized for accuracy in low-complexity genomic regions, we have detected de novo mutations that distinguish nominally identical parasites from clonal lineages. Despite its slower evolutionary rate compared with bacterial or viral species, de novo mutation can be detected in P. falciparum across timescales of just 1-2 years and evolutionary rates in low-complexity regions of the genome can be up to twice that detected in the rest of the genome. The increased mutation rate allows the identification of separate clade expansions that cannot be found using previous genomic epidemiology approaches and could be a crucial tool for mapping residual transmission patterns in disease elimination campaigns and reintroduction scenarios.


Subject(s)
Evolution, Molecular , Malaria/parasitology , Mutation , Plasmodium falciparum/genetics , Genetic Techniques , Malaria/transmission , Phylogeny
11.
Science ; 359(6372): 191-199, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29326268

ABSTRACT

Chemogenetic characterization through in vitro evolution combined with whole-genome analysis can identify antimalarial drug targets and drug-resistance genes. We performed a genome analysis of 262 Plasmodium falciparum parasites resistant to 37 diverse compounds. We found 159 gene amplifications and 148 nonsynonymous changes in 83 genes associated with drug-resistance acquisition, where gene amplifications contributed to one-third of resistance acquisition events. Beyond confirming previously identified multidrug-resistance mechanisms, we discovered hitherto unrecognized drug target-inhibitor pairs, including thymidylate synthase and a benzoquinazolinone, farnesyltransferase and a pyrimidinedione, and a dipeptidylpeptidase and an arylurea. This exploration of the P. falciparum resistome and druggable genome will likely guide drug discovery and structural biology efforts, while also advancing our understanding of resistance mechanisms available to the malaria parasite.


Subject(s)
Antimalarials/pharmacology , Drug Resistance/genetics , Genome, Protozoan , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Activation, Metabolic , Alleles , DNA Copy Number Variations , Directed Molecular Evolution , Drug Resistance, Multiple/genetics , Genes, Protozoan , Metabolomics , Mutation , Plasmodium falciparum/growth & development , Selection, Genetic , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Cell ; 171(7): 1532-1544.e15, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29129376

ABSTRACT

Transmission represents a population bottleneck in the Plasmodium life cycle and a key intervention target of ongoing efforts to eradicate malaria. Sexual differentiation is essential for this process, as only sexual parasites, called gametocytes, are infective to the mosquito vector. Gametocyte production rates vary depending on environmental conditions, but external stimuli remain obscure. Here, we show that the host-derived lipid lysophosphatidylcholine (LysoPC) controls P. falciparum cell fate by repressing parasite sexual differentiation. We demonstrate that exogenous LysoPC drives biosynthesis of the essential membrane component phosphatidylcholine. LysoPC restriction induces a compensatory response, linking parasite metabolism to the activation of sexual-stage-specific transcription and gametocyte formation. Our results reveal that malaria parasites can sense and process host-derived physiological signals to regulate differentiation. These data close a critical knowledge gap in parasite biology and introduce a major component of the sexual differentiation pathway in Plasmodium that may provide new approaches for blocking malaria transmission.


Subject(s)
Lysophosphatidylcholines/metabolism , Malaria/parasitology , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Animals , Female , Humans , Malaria/immunology , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Plasmodium berghei/physiology , Reproduction
13.
Malar J ; 16(1): 195, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28494763

ABSTRACT

BACKGROUND: Artemisinin resistance is associated with delayed parasite clearance half-life in vivo and correlates with ring-stage survival under dihydroartemisinin in vitro. Both phenotypes are associated with mutations in the PF3D7_1343700 pfkelch13 gene. Recent spread of artemisinin resistance and emerging piperaquine resistance in Southeast Asia show that artemisinin combination therapy, such as dihydroartemisinin-piperaquine, are losing clinical effectiveness, prompting investigation of drug resistance mechanisms and development of strategies to surmount emerging anti-malarial resistance. METHODS: Sixty-eight parasites isolates with in vivo clearance data were obtained from two Tracking Resistance to Artemisinin Collaboration study sites in Cambodia, culture-adapted, and genotyped for pfkelch13 and other mutations including pfmdr1 copy number; and the RSA0-3h survival rates and response to antimalarial drugs in vitro were measured for 36 of these isolates. RESULTS: Among these 36 parasites one isolate demonstrated increased ring-stage survival for a PfKelch13 mutation (D584V, RSA0-3h = 8%), previously associated with slow clearance but not yet tested in vitro. Several parasites exhibited increased ring-stage survival, yet lack pfkelch13 mutations, and one isolate showed evidence for piperaquine resistance. CONCLUSIONS: This study of 68 culture-adapted Plasmodium falciparum clinical isolates from Cambodia with known clearance values, associated the D584V PfKelch13 mutation with increased ring-stage survival and identified parasites that lack pfkelch13 mutations yet exhibit increased ring-stage survival. These data suggest mutations other than those found in pfkelch13 may be involved in conferring artemisinin resistance in P. falciparum. Piperaquine resistance was also detected among the same Cambodian samples, consistent with reports of emerging piperaquine resistance in the field. These culture-adapted parasites permit further investigation of mechanisms of both artemisinin and piperaquine resistance and development of strategies to prevent or overcome anti-malarial resistance.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Drug Resistance , Plasmodium falciparum/drug effects , Protozoan Proteins/genetics , Cambodia , Mutation , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism
14.
Mol Microbiol ; 101(3): 381-93, 2016 08.
Article in English | MEDLINE | ID: mdl-27073104

ABSTRACT

Emerging resistance to first-line antimalarial combination therapies threatens malaria treatment and the global elimination campaign. Improved therapeutic strategies are required to protect existing drugs and enhance treatment efficacy. We report that the piperazine-containing compound ACT-451840 exhibits single-digit nanomolar inhibition of the Plasmodium falciparum asexual blood stages and transmissible gametocyte forms. Genome sequence analyses of in vitro-derived ACT-451840-resistant parasites revealed single nucleotide polymorphisms in pfmdr1, which encodes a digestive vacuole membrane-bound ATP-binding cassette transporter known to alter P. falciparum susceptibility to multiple first-line antimalarials. CRISPR-Cas9 based gene editing confirmed that PfMDR1 point mutations mediated ACT-451840 resistance. Resistant parasites demonstrated increased susceptibility to the clinical drugs lumefantrine, mefloquine, quinine and amodiaquine. Stage V gametocytes harboring Cas9-introduced pfmdr1 mutations also acquired ACT-451840 resistance. These findings reveal that PfMDR1 mutations can impart resistance to compounds active against asexual blood stages and mature gametocytes. Exploiting PfMDR1 resistance mechanisms provides new opportunities for developing disease-relieving and transmission-blocking antimalarials.


Subject(s)
Acrylamides/pharmacology , Antimalarials/pharmacology , Artemisinins/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats , Multidrug Resistance-Associated Proteins/metabolism , Piperazines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , DNA, Protozoan/genetics , DNA, Protozoan/metabolism , Drug Resistance , Drug Synergism , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Multidrug Resistance-Associated Proteins/genetics , Plasmodium falciparum/metabolism , Point Mutation , Polymorphism, Single Nucleotide
15.
Sci Transl Med ; 7(288): 288ra77, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995223

ABSTRACT

The emergence of drug resistance is a major limitation of current antimalarials. The discovery of new druggable targets and pathways including those that are critical for multiple life cycle stages of the malaria parasite is a major goal for developing next-generation antimalarial drugs. Using an integrated chemogenomics approach that combined drug resistance selection, whole-genome sequencing, and an orthogonal yeast model, we demonstrate that the cytoplasmic prolyl-tRNA (transfer RNA) synthetase (PfcPRS) of the malaria parasite Plasmodium falciparum is a biochemical and functional target of febrifugine and its synthetic derivative halofuginone. Febrifugine is the active principle of a traditional Chinese herbal remedy for malaria. We show that treatment with febrifugine derivatives activated the amino acid starvation response in both P. falciparum and a transgenic yeast strain expressing PfcPRS. We further demonstrate in the Plasmodium berghei mouse model of malaria that halofuginol, a new halofuginone analog that we developed, is active against both liver and asexual blood stages of the malaria parasite. Halofuginol, unlike halofuginone and febrifugine, is well tolerated at efficacious doses and represents a promising lead for the development of dual-stage next-generation antimalarials.


Subject(s)
Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Malaria, Falciparum/drug therapy , Piperidines/pharmacology , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Quinazolinones/pharmacology , Amino Acyl-tRNA Synthetases/metabolism , Animals , Antimalarials/chemistry , Antimalarials/toxicity , Computer-Aided Design , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Drug Resistance , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Erythrocytes/parasitology , Liver/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Mice , Models, Molecular , Molecular Structure , Molecular Targeted Therapy , Piperidines/chemistry , Piperidines/toxicity , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Quinazolines/chemistry , Quinazolines/toxicity , Quinazolinones/chemistry , Quinazolinones/toxicity , Structure-Activity Relationship , Time Factors
16.
BMC Bioinformatics ; 15: 63, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24589256

ABSTRACT

BACKGROUND: Whole-genome sequencing represents a powerful experimental tool for pathogen research. We present methods for the analysis of small eukaryotic genomes, including a streamlined system (called Platypus) for finding single nucleotide and copy number variants as well as recombination events. RESULTS: We have validated our pipeline using four sets of Plasmodium falciparum drug resistant data containing 26 clones from 3D7 and Dd2 background strains, identifying an average of 11 single nucleotide variants per clone. We also identify 8 copy number variants with contributions to resistance, and report for the first time that all analyzed amplification events are in tandem. CONCLUSIONS: The Platypus pipeline provides malaria researchers with a powerful tool to analyze short read sequencing data. It provides an accurate way to detect SNVs using known software packages, and a novel methodology for detection of CNVs, though it does not currently support detection of small indels. We have validated that the pipeline detects known SNVs in a variety of samples while filtering out spurious data. We bundle the methods into a freely available package.


Subject(s)
DNA Copy Number Variations/genetics , Genome, Protozoan/genetics , Genomics/methods , Plasmodium falciparum/genetics , Software , Antimalarials/pharmacology , DNA, Protozoan/genetics , Drug Resistance/genetics , Plasmodium falciparum/drug effects , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA/methods
17.
Malar J ; 12: 316, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24025732

ABSTRACT

BACKGROUND: One of the main causes of mortality from severe malaria in Plasmodium falciparum infections is cerebral malaria (CM). An important host genetic component determines the susceptibility of an individual to develop CM or to clear the infection and become semi-immune. As such, the identification of genetic loci associated with susceptibility or resistance may serve to modulate disease severity. METHODOLOGY: The Plasmodium berghei mouse model for experimental cerebral malaria (ECM) reproduces several disease symptoms seen in human CM, and two different phenotypes, a susceptible (FVB/NJ) and a resistant mouse strain (DBA/2J), were examined. RESULTS: FVB/NJ mice died from infection within ten days, whereas DBA/2J mice showed a gender bias: males survived on average nineteen days and females either died early with signs of ECM or survived for up to three weeks. A comparison of brain pathology between FVB/NJ and DBA/2J showed no major differences with regard to brain haemorrhages or the number of parasites and CD3+ cells in the microvasculature. However, significant differences were found in the peripheral blood of infected mice: For example resistant DBA/2J mice had significantly higher numbers of circulating basophils than did FVB/NJ mice on day seven. Analysis of the F2 offspring from a cross of DBA/2J and FVB/NJ mice mapped the genetic locus of the underlying survival trait to chromosome 9 with a Lod score of 4.9. This locus overlaps with two previously identified resistance loci (char1 and pymr) from a blood stage malaria model. CONCLUSIONS: Survival best distinguishes malaria infections between FVB/NJ and DBA/2J mice. The importance of char1 and pymr on chromosome 9 in malaria resistance to P. berghei was confirmed. In addition there was an association of basophil numbers with survival.


Subject(s)
Chromosomes, Human, Pair 9 , Disease Resistance , Genetic Loci , Malaria, Cerebral/genetics , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Animals , Basophils/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Survival Analysis
18.
PLoS Genet ; 9(2): e1003293, 2013.
Article in English | MEDLINE | ID: mdl-23408914

ABSTRACT

Malaria parasites elude eradication attempts both within the human host and across nations. At the individual level, parasites evade the host immune responses through antigenic variation. At the global level, parasites escape drug pressure through single nucleotide variants and gene copy amplification events conferring drug resistance. Despite their importance to global health, the rates at which these genomic alterations emerge have not been determined. We studied the complete genomes of different Plasmodium falciparum clones that had been propagated asexually over one year in the presence and absence of drug pressure. A combination of whole-genome microarray analysis and next-generation deep resequencing (totaling 14 terabases) revealed a stable core genome with only 38 novel single nucleotide variants appearing in seventeen evolved clones (avg. 5.4 per clone). In clones exposed to atovaquone, we found cytochrome b mutations as well as an amplification event encompassing the P. falciparum multidrug resistance associated protein (mrp1) on chromosome 1. We observed 18 large-scale (>1 kb on average) deletions of telomere-proximal regions encoding multigene families, involved in immune evasion (9.5×10(-6) structural variants per base pair per generation). Six of these deletions were associated with chromosomal crossovers generated during mitosis. We found only minor differences in rates between genetically distinct strains and between parasites cultured in the presence or absence of drug. Using these derived mutation rates for P. falciparum (1.0-9.7×10(-9) mutations per base pair per generation), we can now model the frequency at which drug or immune resistance alleles will emerge under a well-defined set of assumptions. Further, the detection of mitotic recombination events in var gene families illustrates how multigene families can arise and change over time in P. falciparum. These results will help improve our understanding of how P. falciparum evolves to evade control efforts within both the individual hosts and large populations.


Subject(s)
Antigens , Atovaquone/administration & dosage , Drug Resistance, Multiple , Host-Parasite Interactions , Plasmodium falciparum , Antigenic Variation/drug effects , Antigenic Variation/genetics , Antigens/drug effects , Antigens/genetics , Cytochromes b/genetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Evolution, Molecular , Genome, Protozoan/drug effects , High-Throughput Nucleotide Sequencing , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Mitosis/genetics , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/immunology , Multidrug Resistance-Associated Proteins/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology
19.
J Am Chem Soc ; 134(41): 17320-32, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-22978674

ABSTRACT

An improved sulfenylation method for the preparation of epidithio-, epitetrathio-, and bis-(methylthio)diketopiperazines from diketopiperazines has been developed. Employing NaHMDS and related bases and elemental sulfur or bis[bis(trimethylsilyl)amino]trisulfide (23) in THF, the developed method was applied to the synthesis of a series of natural and designed molecules, including epicoccin G (1), 8,8'-epi-ent-rostratin B (2), gliotoxin (3), gliotoxin G (4), emethallicin E (5), and haematocin (6). Biological screening of selected synthesized compounds led to the discovery of a number of nanomolar antipoliovirus agents (i.e., 46, 2,2'-epi-46, and 61) and several low-micromolar anti- Plasmodium falciparum lead compounds (i.e., 46, 2,2'-epi-46, 58, 61, and 1).


Subject(s)
Antimalarials/pharmacology , Antiviral Agents/pharmacology , Diketopiperazines/pharmacology , Plasmodium falciparum/drug effects , Poliovirus/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Diketopiperazines/chemical synthesis , Diketopiperazines/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Conformation , Parasitic Sensitivity Tests , Stereoisomerism , Structure-Activity Relationship
20.
Science ; 334(6061): 1372-7, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22096101

ABSTRACT

Most malaria drug development focuses on parasite stages detected in red blood cells, even though, to achieve eradication, next-generation drugs active against both erythrocytic and exo-erythrocytic forms would be preferable. We applied a multifactorial approach to a set of >4000 commercially available compounds with previously demonstrated blood-stage activity (median inhibitory concentration < 1 micromolar) and identified chemical scaffolds with potent activity against both forms. From this screen, we identified an imidazolopiperazine scaffold series that was highly enriched among compounds active against Plasmodium liver stages. The orally bioavailable lead imidazolopiperazine confers complete causal prophylactic protection (15 milligrams/kilogram) in rodent models of malaria and shows potent in vivo blood-stage therapeutic activity. The open-source chemical tools resulting from our effort provide starting points for future drug discovery programs, as well as opportunities for researchers to investigate the biology of exo-erythrocytic forms.


Subject(s)
Antimalarials/pharmacology , Drug Discovery , Imidazoles/pharmacology , Liver/parasitology , Malaria/drug therapy , Piperazines/pharmacology , Plasmodium/drug effects , Animals , Antimalarials/chemistry , Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Resistance , Erythrocytes/parasitology , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Molecular Structure , Piperazines/chemistry , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Plasmodium/cytology , Plasmodium/growth & development , Plasmodium/physiology , Plasmodium berghei/cytology , Plasmodium berghei/drug effects , Plasmodium berghei/growth & development , Plasmodium berghei/physiology , Plasmodium falciparum/cytology , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Plasmodium falciparum/physiology , Plasmodium yoelii/cytology , Plasmodium yoelii/drug effects , Plasmodium yoelii/growth & development , Plasmodium yoelii/physiology , Polymorphism, Single Nucleotide , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Random Allocation , Small Molecule Libraries , Sporozoites/drug effects , Sporozoites/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL