Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
BMC Genomics ; 23(1): 780, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36451097

ABSTRACT

BACKGROUND: Important regulation occurs at the level of transcription in Plasmodium falciparum and growing evidence suggests that these apicomplexan parasites have complex regulatory networks. Recent studies implicate long noncoding RNAs (lncRNAs) as transcriptional regulators in P. falciparum. However, due to limited research and the lack of necessary experimental tools, our understanding of their role in the malaria-causing parasite remains largely unelucidated. In this work, we address one of these limitations, the lack of an updated and improved lncRNA annotation in P. falciparum. RESULTS: We generated long-read RNA sequencing data and integrated information extracted and curated from multiple sources to manually annotate lncRNAs. We identified 1119 novel lncRNAs and validated and refined 1250 existing annotations. Utilising the collated datasets, we generated evidence-based ranking scores for each annotation and characterised the distinct genomic contexts and features of P. falciparum lncRNAs. Certain features indicated subsets with potential biological significance such as 25 lncRNAs containing multiple introns, 335 lncRNAs lacking mutations in piggyBac mutagenic studies and lncRNAs associated with specific biologic processes including two new types of lncRNAs found proximal to var genes. CONCLUSIONS: The insights and the annotation presented in this study will serve as valuable tools for researchers seeking to understand the role of lncRNAs in parasite biology through both bioinformatics and experimental approaches.


Subject(s)
Malaria, Falciparum , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Genomics , Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Computational Biology
3.
Methods Mol Biol ; 2470: 221-239, 2022.
Article in English | MEDLINE | ID: mdl-35881349

ABSTRACT

The ability to interrogate gene function in Plasmodium parasites has been greatly enhanced by the advent of CRISPR/Cas9 systems. The breadth of genome manipulations ranges from single point mutations to large multigene deletions, however many of the technical considerations for designing CRISPR-based experiments are common to any editing approach. This review will discuss protocols for vector construction and donor design for genome editing P. falciparum, including pitfalls, variables, and validation methods.


Subject(s)
Plasmodium falciparum , Plasmodium , CRISPR-Cas Systems/genetics , Gene Editing/methods , Genome, Protozoan , Plasmodium falciparum/genetics
4.
PLoS Pathog ; 17(7): e1008864, 2021 07.
Article in English | MEDLINE | ID: mdl-34197567

ABSTRACT

Plasmodium vivax is responsible for the majority of malaria cases outside Africa. Unlike P. falciparum, the P. vivax life-cycle includes a dormant liver stage, the hypnozoite, which can cause infection in the absence of mosquito transmission. An effective vaccine against P. vivax blood stages would limit symptoms and pathology from such recurrent infections, and therefore could play a critical role in the control of this species. Vaccine development in P. vivax, however, lags considerably behind P. falciparum, which has many identified targets with several having transitioned to Phase II testing. By contrast only one P. vivax blood-stage vaccine candidate based on the Duffy Binding Protein (PvDBP), has reached Phase Ia, in large part because the lack of a continuous in vitro culture system for P. vivax limits systematic screening of new candidates. We used the close phylogenetic relationship between P. vivax and P. knowlesi, for which an in vitro culture system in human erythrocytes exists, to test the scalability of systematic reverse vaccinology to identify and prioritise P. vivax blood-stage targets. A panel of P. vivax proteins predicted to function in erythrocyte invasion were expressed as full-length recombinant ectodomains in a mammalian expression system. Eight of these antigens were used to generate polyclonal antibodies, which were screened for their ability to recognize orthologous proteins in P. knowlesi. These antibodies were then tested for inhibition of growth and invasion of both wild type P. knowlesi and chimeric P. knowlesi lines modified using CRISPR/Cas9 to exchange P. knowlesi genes with their P. vivax orthologues. Candidates that induced antibodies that inhibited invasion to a similar level as PvDBP were identified, confirming the utility of P. knowlesi as a model for P. vivax vaccine development and prioritizing antigens for further follow up.


Subject(s)
Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium knowlesi/immunology , Plasmodium vivax/immunology , Antigens, Protozoan/immunology , Cells, Cultured , Humans , Malaria, Vivax/prevention & control , Protozoan Proteins/immunology
5.
Sci Rep ; 10(1): 10894, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616799

ABSTRACT

The recurrent emergence of drug resistance in Plasmodium falciparum increases the urgency to genetically validate drug resistance mechanisms and identify new targets. Reverse genetics have facilitated genome-scale knockout screens in Plasmodium berghei and Toxoplasma gondii, in which pooled transfections of multiple vectors were critical to increasing scale and throughput. These approaches have not yet been implemented in human malaria species such as P. falciparum and P. knowlesi, in part because the extent to which pooled transfections can be performed in these species remains to be evaluated. Here we use next-generation sequencing to quantitate uptake of a pool of 94 barcoded vectors. The distribution of vector acquisition allowed us to estimate the number of barcodes and DNA molecules taken up by the parasite population. Dilution cloning of P. falciparum transfectants showed that individual clones possess as many as seven episomal barcodes, revealing that an intake of multiple vectors is a frequent event despite the inefficient transfection efficiency. Transfection of three spectrally-distinct fluorescent reporters allowed us to evaluate different transfection methods and revealed that schizont-stage transfection limited the tendency for parasites to take up multiple vectors. In contrast to P. falciparum, we observed that the higher transfection efficiency of P. knowlesi resulted in near complete representation of the library. These findings have important implications for how reverse genetics can be scaled in culturable Plasmodium species.


Subject(s)
DNA, Recombinant/metabolism , Genetic Vectors/metabolism , Plasmids/metabolism , Plasmodium falciparum/metabolism , Transfection/methods , Biological Transport , Calmodulin/genetics , Clone Cells , DNA Barcoding, Taxonomic , Electroporation , Erythrocytes/parasitology , Flow Cytometry , Gene Library , Genetic Vectors/genetics , Humans , Luminescent Proteins/genetics , Plasmids/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium knowlesi/genetics , Plasmodium knowlesi/growth & development , Plasmodium knowlesi/metabolism , Promoter Regions, Genetic , Species Specificity
6.
Genome Res ; 29(12): 1974-1984, 2019 12.
Article in English | MEDLINE | ID: mdl-31740578

ABSTRACT

Cryptic transcription is widespread and generates a heterogeneous group of RNA molecules of unknown function. To improve our understanding of cryptic transcription, we investigated their transcription start site (TSS) usage, chromatin organization, and posttranscriptional consequences in Saccharomyces cerevisiae We show that TSSs of chromatin-sensitive internal cryptic transcripts retain comparable features of canonical TSSs in terms of DNA sequence, directionality, and chromatin accessibility. We define the 5' and 3' boundaries of cryptic transcripts and show that, contrary to RNA degradation-sensitive ones, they often overlap with the end of the gene, thereby using the canonical polyadenylation site, and associate to polyribosomes. We show that chromatin-sensitive cryptic transcripts can be recognized by ribosomes and may produce truncated polypeptides from downstream, in-frame start codons. Finally, we confirm the presence of the predicted polypeptides by reanalyzing N-terminal proteomic data sets. Our work suggests that a fraction of chromatin-sensitive internal cryptic promoters initiates the transcription of alternative truncated mRNA isoforms. The expression of these chromatin-sensitive isoforms is conserved from yeast to human, expanding the functional consequences of cryptic transcription and proteome complexity.


Subject(s)
Chromatin , Gene Expression Regulation, Fungal , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Initiation Site , Chromatin/genetics , Chromatin/metabolism , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Stability , RNA, Fungal/biosynthesis , RNA, Fungal/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
7.
PLoS Pathog ; 15(10): e1008086, 2019 10.
Article in English | MEDLINE | ID: mdl-31658303

ABSTRACT

Plasmodium parasites are the causative agents of malaria, a disease with wide public health repercussions. Increasing drug resistance and the absence of a vaccine make finding new chemotherapeutic strategies imperative. Components of the ubiquitin and ubiquitin-like pathways have garnered increased attention as novel targets given their necessity to parasite survival. Understanding how these pathways are regulated in Plasmodium and identifying differences to the host is paramount to selectively interfering with parasites. Here, we focus on Nedd8 modification in Plasmodium falciparum, given its central role to cell division and DNA repair, processes critical to Plasmodium parasites given their unusual cell cycle and requirement for refined repair mechanisms. By applying a functional chemical approach, we show that deNeddylation is controlled by a different set of enzymes in the parasite versus the human host. We elucidate the molecular determinants of the unusual dual ubiquitin/Nedd8 recognition by the essential PfUCH37 enzyme and, through parasite transgenics and drug assays, determine that only its ubiquitin activity is critical to parasite survival. Our experiments reveal interesting evolutionary differences in how neddylation is controlled in higher versus lower eukaryotes, and highlight the Nedd8 pathway as worthy of further exploration for therapeutic targeting in antimalarial drug design.


Subject(s)
NEDD8 Protein/metabolism , Plasmodium falciparum/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Antimalarials/pharmacology , Cell Line , HEK293 Cells , Humans , Hydrolysis , Malaria, Falciparum/drug therapy , Malaria, Falciparum/pathology , Ubiquitination/physiology
8.
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
9.
Methods Mol Biol ; 1689: 177-194, 2018.
Article in English | MEDLINE | ID: mdl-29027175

ABSTRACT

Chromatin immunoprecipitation followed by sequencing (ChIP-Seq) or microarray hybridization (ChIP-on-chip) are standard methods for the study of transcription factor binding sites and histone chemical modifications. However, these approaches only allow profiling of a single factor or protein modification at a time.In this chapter, we present Bar-ChIP, a higher throughput version of ChIP-Seq that relies on the direct ligation of molecular barcodes to chromatin fragments. Bar-ChIP enables the concurrent profiling of multiple DNA-protein interactions and is therefore amenable to experimental scale-up, without the need for any robotic instrumentation.


Subject(s)
Chromatin Immunoprecipitation , DNA Barcoding, Taxonomic , High-Throughput Nucleotide Sequencing , Nucleosomes/genetics , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation/methods , Computational Biology/methods , Data Interpretation, Statistical , Gene Library , High-Throughput Nucleotide Sequencing/methods , Nucleosomes/metabolism , Real-Time Polymerase Chain Reaction/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Workflow
10.
Nat Commun ; 7: 13401, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27824335

ABSTRACT

Plasmodium falciparum infections can cause severe malaria, but not every infected person develops life-threatening complications. In particular, carriers of the structural haemoglobinopathies S and C and infants are protected from severe disease. Protection is associated with impaired parasite-induced host actin reorganization, required for vesicular trafficking of parasite-encoded adhesins, and reduced cytoadherence of parasitized erythrocytes in the microvasculature. Here we show that aberrant host actin remodelling and the ensuing reduced cytoadherence result from a redox imbalance inherent to haemoglobinopathic and fetal erythrocytes. We further show that a transient oxidative insult to wild-type erythrocytes before infection with P. falciparum induces the phenotypic features associated with the protective trait of haemoglobinopathic and fetal erythrocytes. Moreover, pretreatment of mice with the pro-oxidative nutritional supplement menadione mitigate the development of experimental cerebral malaria. Our results identify redox imbalance as a causative principle of protection from severe malaria, which might inspire host-directed intervention strategies.


Subject(s)
Anemia, Sickle Cell/blood , Erythrocytes/parasitology , Fetus/pathology , Malaria, Falciparum/pathology , Malaria, Falciparum/parasitology , Oxidative Stress , Actins/metabolism , Animals , Cytoplasm/metabolism , Erythrocytes/ultrastructure , Female , Hemoglobins/metabolism , Mice, Inbred C57BL , Models, Biological , Oxidation-Reduction , Phenotype , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Plasmodium falciparum/metabolism , Plasmodium falciparum/ultrastructure , Vitamin K 3/pharmacology
11.
Antimicrob Agents Chemother ; 60(9): 5146-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27297478

ABSTRACT

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


Subject(s)
Antimalarials/pharmacology , Gametogenesis/drug effects , Life Cycle Stages/drug effects , Naphthoquinones/pharmacology , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Artemisinins/pharmacology , Atovaquone/pharmacology , Drug Interactions , Drug Resistance/drug effects , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Inhibitory Concentration 50 , Methylene Blue/pharmacology , Naphthoquinones/chemical synthesis , Plasmodium falciparum/growth & development
12.
Cell Rep ; 14(10): 2463-75, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947071

ABSTRACT

A comprehensive map of transcription start sites (TSSs) across the highly AT-rich genome of P. falciparum would aid progress toward deciphering the molecular mechanisms that underlie the timely regulation of gene expression in this malaria parasite. Using high-throughput sequencing technologies, we generated a comprehensive atlas of transcription initiation events at single-nucleotide resolution during the parasite intra-erythrocytic developmental cycle. This detailed analysis of TSS usage enabled us to define architectural features of plasmodial promoters. We demonstrate that TSS selection and strength are constrained by local nucleotide composition. Furthermore, we provide evidence for coordinate and stage-specific TSS usage from distinct sites within the same transcription unit, thereby producing transcript isoforms, a subset of which are developmentally regulated. This work offers a framework for further investigations into the interactions between genomic sequences and regulatory factors governing the complex transcriptional program of this major human pathogen.


Subject(s)
Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Transcription, Genetic , 5' Untranslated Regions , Blotting, Northern , Humans , Life Cycle Stages/genetics , Malaria/parasitology , Plasmodium falciparum/growth & development , Promoter Regions, Genetic , Protein Isoforms , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Regulatory Elements, Transcriptional/genetics , Transcription Initiation Site
13.
BMC Genomics ; 16: 1090, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26689807

ABSTRACT

BACKGROUND: The acquisition of multidrug resistance by Plasmodium falciparum underscores the need to understand the underlying molecular mechanisms so as to counter their impact on malaria control. For the many antimalarials whose mode of action relates to inhibition of heme detoxification inside infected erythrocytes, the digestive vacuole transporters PfCRT and PfMDR1 constitute primary resistance determinants. RESULTS: Using gene expression microarrays over the course of the parasite intra-erythrocytic developmental cycle, we compared the transcriptomic profiles between P. falciparum strains displaying mutant or wild-type pfcrt or varying in pfcrt or pfmdr1 expression levels. To account for differences in the time of sampling, we developed a computational method termed Hypergeometric Analysis of Time Series, which combines Fast Fourier Transform with a modified Gene Set Enrichment Analysis. Our analysis revealed coordinated changes in genes involved in protein catabolism, translation initiation and DNA/RNA metabolism. We also observed differential expression of genes with a role in transport or coding for components of the digestive vacuole. Interestingly, a global comparison of all profiled transcriptomes uncovered a tight correlation between the transcript levels of pfcrt and pfmdr1, extending to dozens of other genes, suggesting an intricate regulatory balance in order to maintain optimal physiological processes. CONCLUSIONS: This study provides insight into the mechanisms by which P. falciparum adjusts to the acquisition of mutations or gene amplification in key transporter loci that mediate drug resistance. Our results implicate several biological pathways that may be differentially regulated to compensate for impaired transporter function and alterations in parasite vacuole physiology.


Subject(s)
Gene Expression Profiling/methods , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Computational Biology/methods , Drug Resistance, Multiple , Gene Amplification , Gene Expression Regulation , Mutation , Plasmodium falciparum/physiology
14.
ACS Chem Biol ; 10(3): 775-83, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25602169

ABSTRACT

Equilibrative transporters are potential drug targets; however, most functional assays involve radioactive substrate uptake that is unsuitable for high-throughput screens (HTS). We developed a robust yeast-based growth assay that is potentially applicable to many equilibrative transporters. As proof of principle, we applied our approach to Equilibrative Nucleoside Transporter 1 of the malarial parasite Plasmodium falciparum (PfENT1). PfENT1 inhibitors might serve as novel antimalarial drugs since PfENT1-mediated purine import is essential for parasite proliferation. To identify PfENT1 inhibitors, we screened 64 560 compounds and identified 171 by their ability to rescue the growth of PfENT1-expressing fui1Δ yeast in the presence of a cytotoxic PfENT1 substrate, 5-fluorouridine (5-FUrd). In secondary assays, nine of the highest activity compounds inhibited PfENT1-dependent growth of a purine auxotrophic yeast strain with adenosine as the sole purine source (IC50 0.2-2 µM). These nine compounds completely blocked [(3)H]adenosine uptake into PfENT1-expressing yeast and erythrocyte-free trophozoite-stage parasites (IC50 5-50 nM), and inhibited chloroquine-sensitive and -resistant parasite proliferation (IC50 5-50 µM). Wild-type (WT) parasite IC50 values were up to 4-fold lower compared to PfENT1-knockout (pfent1Δ) parasites. pfent1Δ parasite killing showed a delayed-death phenotype not observed with WT. We infer that, in parasites, the compounds inhibit both PfENT1 and a secondary target with similar efficacy. The secondary target identity is unknown, but its existence may reduce the likelihood of parasites developing resistance to PfENT1 inhibitors. Our data support the hypothesis that blocking purine transport through PfENT1 may be a novel and compelling approach for antimalarial drug development.


Subject(s)
Antimalarials/pharmacology , High-Throughput Screening Assays , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/antagonists & inhibitors , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Trophozoites/drug effects , Adenosine/metabolism , Antimalarials/chemistry , Axenic Culture , Biological Transport/drug effects , Gene Deletion , Gene Expression , Genetic Complementation Test , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/genetics , Nucleobase, Nucleoside, Nucleotide, and Nucleic Acid Transport Proteins/metabolism , Nucleoside Transport Proteins/genetics , Nucleoside Transport Proteins/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Trophozoites/growth & development , Trophozoites/metabolism , Uridine/analogs & derivatives , Uridine/pharmacology
15.
Mol Syst Biol ; 11(1): 777, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25583149

ABSTRACT

We present a modified approach of chromatin immuno-precipitation followed by sequencing (ChIP-Seq), which relies on the direct ligation of molecular barcodes to chromatin fragments, thereby permitting experimental scale-up. With Bar-ChIP now enabling the concurrent profiling of multiple DNA-protein interactions, we report the simultaneous generation of 90 ChIP-Seq datasets without any robotic instrumentation. We demonstrate that application of Bar-ChIP to a panel of Saccharomyces cerevisiae chromatin-associated mutants provides a rapid and accurate genome-wide overview of their chromatin status. Additionally, we validate the utility of this technology to derive novel biological insights by identifying a role for the Rpd3S complex in maintaining H3K14 hypo-acetylation in gene bodies. We also report an association between the presence of intragenic H3K4 tri-methylation and the emergence of cryptic transcription in a Set2 mutant. Finally, we uncover a crosstalk between H3K14 acetylation and H3K4 methylation in this mutant. These results show that Bar-ChIP enables biological discovery through rapid chromatin profiling at single-nucleosome resolution for various conditions and protein modifications at once.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/genetics , High-Throughput Nucleotide Sequencing/methods , Acetylation , Chromatin/chemistry , DNA, Fungal/genetics , Databases, Genetic , Gene Expression Profiling , Genetic Association Studies , Genetic Markers , Histones/genetics , Histones/metabolism , Methylation , Nucleosomes , Reproducibility of Results , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Analysis, DNA
16.
Proc Natl Acad Sci U S A ; 110(45): 18262-7, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24145416

ABSTRACT

Efficient transmission of Plasmodium species between humans and Anopheles mosquitoes is a major contributor to the global burden of malaria. Gametocytogenesis, the process by which parasites switch from asexual replication within human erythrocytes to produce male and female gametocytes, is a critical step in malaria transmission and Plasmodium genetic diversity. Nothing is known about the pathways that regulate gametocytogenesis and only few of the current drugs that inhibit asexual replication are also capable of inhibiting gametocyte development and blocking malaria transmission. Here we provide genetic and pharmacological evidence indicating that the pathway for synthesis of phosphatidylcholine in Plasmodium falciparum membranes from host serine is essential for parasite gametocytogenesis and malaria transmission. Parasites lacking the phosphoethanolamine N-methyltransferase enzyme, which catalyzes the limiting step in this pathway, are severely altered in gametocyte development, are incapable of producing mature-stage gametocytes, and are not transmitted to mosquitoes. Chemical screening identified 11 inhibitors of phosphoethanolamine N-methyltransferase that block parasite intraerythrocytic asexual replication and gametocyte differentiation in the low micromolar range. Kinetic studies in vitro as well as functional complementation assays and lipid metabolic analyses in vivo on the most promising inhibitor NSC-158011 further demonstrated the specificity of inhibition. These studies set the stage for further optimization of NSC-158011 for development of a class of dual activity antimalarials to block both intraerythrocytic asexual replication and gametocytogenesis.


Subject(s)
Enzyme Inhibitors/pharmacology , Malaria, Falciparum/transmission , Methyltransferases/metabolism , Plasmodium falciparum/enzymology , Reproduction, Asexual/drug effects , Antimalarials/pharmacology , Female , Fluorescent Antibody Technique , Humans , Malaria, Falciparum/enzymology , Male , Methyltransferases/antagonists & inhibitors , Plasmodium falciparum/growth & development , Radiometry , Serine/metabolism
17.
Antimicrob Agents Chemother ; 57(12): 6050-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24060871

ABSTRACT

The design of new antimalarial combinations to treat Plasmodium falciparum infections requires drugs that, in addition to resolving disease symptoms caused by asexual blood stage parasites, can also interrupt transmission to the mosquito vector. Gametocytes, which are essential for transmission, develop as sexual blood stage parasites in the human host over 8 to 12 days and are the most accessible developmental stage for transmission-blocking drugs. Considerable effort is currently being devoted to identifying compounds active against mature gametocytes. However, investigations on the drug sensitivity of developing gametocytes, as well as screening methods for identifying inhibitors of early gametocytogenesis, remain scarce. We have developed a luciferase-based high-throughput screening (HTS) assay using tightly synchronous stage I to III gametocytes from a recombinant P. falciparum line expressing green fluorescent protein (GFP)-luciferase. The assay has been used to evaluate the early-stage gametocytocidal activity of the MMV Malaria Box, a collection of 400 compounds with known antimalarial (asexual blood stage) activity. Screening this collection against early-stage (I to III) gametocytes yielded 64 gametocytocidal compounds with 50% inhibitory concentrations (IC50s) below 2.5 µM. This assay is reproducible and suitable for the screening of large compound libraries, with an average percent coefficient of variance (%CV) of ≤5%, an average signal-to-noise ratio (S:N) of >30, and a Z' of ∼0.8. Our findings highlight the need for screening efforts directed specifically against early gametocytogenesis and indicate the importance of experimental verification of early-stage gametocytocidal activity in the development of new antimalarial candidates for combination therapy.


Subject(s)
Antigens, Protozoan/genetics , Antimalarials/pharmacology , High-Throughput Screening Assays/methods , Life Cycle Stages/drug effects , Membrane Proteins/genetics , Plasmodium falciparum/drug effects , Antigens, Protozoan/metabolism , Antimalarials/chemistry , Databases, Chemical , Erythrocytes/drug effects , Erythrocytes/parasitology , Gene Expression , Genes, Reporter , High-Throughput Screening Assays/standards , Humans , Inhibitory Concentration 50 , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/metabolism , Parasitic Sensitivity Tests , Plasmodium falciparum/growth & development , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 108(47): E1214-23, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22042867

ABSTRACT

Clinical studies and mathematical models predict that, to achieve malaria elimination, combination therapies will need to incorporate drugs that block the transmission of Plasmodium falciparum sexual stage parasites to mosquito vectors. Efforts to measure the activity of existing antimalarials on intraerythrocytic sexual stage gametocytes and identify transmission-blocking agents have, until now, been hindered by a lack of quantitative assays. Here, we report an experimental system using P. falciparum lines that stably express gametocyte-specific GFP-luciferase reporters, which enable the assessment of dose- and time-dependent drug action on gametocyte maturation and transmission. These studies reveal activity of the first-line antimalarial dihydroartemisinin and the partner drugs lumefantrine and pyronaridine against early gametocyte stages, along with moderate inhibition of mature gametocyte transmission to Anopheles mosquitoes. The other partner agents monodesethyl-amodiaquine and piperaquine showed activity only against immature gametocytes. Our data also identify methylene blue as a potent inhibitor of gametocyte development across all stages. This thiazine dye almost fully abolishes P. falciparum transmission to mosquitoes at concentrations readily achievable in humans, highlighting the potential of this chemical class to reduce the spread of malaria.


Subject(s)
Anopheles/microbiology , Antimalarials/pharmacology , Malaria/transmission , Methylene Blue/pharmacology , Plasmodium falciparum/physiology , Sexual Development/physiology , Amodiaquine/analogs & derivatives , Animals , Artemisinins , Blotting, Southern , Dose-Response Relationship, Drug , Ethanolamines , Fluorenes , Genetic Vectors , Germ Cells, Plant/drug effects , Green Fluorescent Proteins , Luciferases , Lumefantrine , Naphthyridines , Plasmodium falciparum/drug effects , Quinolines
19.
Methods Mol Biol ; 634: 87-100, 2010.
Article in English | MEDLINE | ID: mdl-20676977

ABSTRACT

Genetic manipulation of the human malaria parasite Plasmodium falciparum has presented substantial challenges for research efforts aimed at better understanding the complex biology of this highly virulent organism. The development of methods to perform gene disruption, allelic replacement or transgene expression has provided important insights into the function of parasite genes. However, genomic integration studies have been hindered by low transfection and recombination efficiencies, and are complicated by the propensity of this parasite to maintain episomal replicating plasmids. We have developed a fast and efficient site-specific system of integrative recombination into the P. falciparum genome, which is catalyzed by the mycobacteriophage Bxb1 serine integrase. This system has the advantage of providing greater genetic and phenotypic homogeneity within transgenic lines as compared to earlier methods based on episomal replication of plasmids. Herein, we present this methodology.


Subject(s)
Genes, Protozoan , Integrases/metabolism , Mycobacteriophages/enzymology , Plasmodium falciparum/genetics , Animals , Base Sequence , DNA Primers , Plasmids , Recombination, Genetic
20.
PLoS One ; 4(9): e6889, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19707292

ABSTRACT

BACKGROUND: Plasmodium falciparum parasitization of erythrocytes causes a substantial increase in the levels of intracellular fatty acids, notably oleic acid. How parasites acquire this monounsaturated fatty acid has remained enigmatic. Here, we report on the biochemical and enzymatic characterization of stearoyl-CoA desaturase (SCD) in P. falciparum. METHODOLOGY/PRINCIPAL FINDINGS: Metabolic labeling experiments allowed us to demonstrate the production of oleic acid from stearic acid both in lysates of parasites incubated with [(14)C]-stearoyl-CoA and in parasite-infected erythrocytes labeled with [(14)C]-stearic acid. Optimal SCD activity was detected in schizonts, the stage of maximal membrane synthesis. This activity correlated with a late trophozoite stage-specific induction of PFE0555w transcripts. PFE0555w harbors a typical SCD signature. Similar to mammalian SCDs, this protein was found to be associated with the endoplasmic reticulum, as determined with PFE0555w-GFP tagged transgenic P. falciparum. Importantly, these parasites exhibited increased rates of stearic to oleic acid conversion, providing additional evidence that PFE0555w encodes the plasmodial SCD (PfSCD). These findings prompted us to assess the activity of sterculic acid analogues, known to be specific Delta9-desaturase inhibitors. Methyl sterculate inhibited the synthesis of oleic acid both with parasite lysates and infected erythrocytes, most likely by targeting PfSCD. This compound exhibited significant, rapid and irreversible antimalarial activity against asexual blood stages. This parasiticidal effect was antagonized by oleic acid. CONCLUSION/SIGNIFICANCE: Our study provides evidence that parasite-mediated fatty acid modification is important for blood-stage survival and provides a new strategy to develop a novel antimalarial therapeutic based on the inhibition of PfSCD.


Subject(s)
Antimalarials/therapeutic use , Oleic Acid/biosynthesis , Plasmodium falciparum/metabolism , Stearoyl-CoA Desaturase/chemistry , Stearoyl-CoA Desaturase/therapeutic use , Amino Acid Sequence , Animals , Animals, Genetically Modified , Antimalarials/chemistry , Cyclopropanes/chemistry , Erythrocytes/parasitology , Escherichia coli/metabolism , Fatty Acids/chemistry , Green Fluorescent Proteins/metabolism , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...