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










Publication year range
1.
Dis Model Mech ; 16(2)2023 02 01.
Article in English | MEDLINE | ID: mdl-36715290

ABSTRACT

Phenotypic cell-based screens are critical tools for discovering candidate drugs for development, yet identification of the cellular target and mode of action of a candidate drug is often lacking. Using an imaging-based screen, we recently discovered an N-[(4-hydroxychroman-4-yl)methyl]-sulphonamide (N-4HCS) compound, DDD01035881, that blocks male gamete formation in the malaria parasite life cycle and subsequent transmission of the parasite to the mosquito with nanomolar activity. To identify the target(s) of DDD01035881, and of the N-4HCS class of compounds more broadly, we synthesised a photoactivatable derivative, probe 2. Photoaffinity labelling of probe 2 coupled with mass spectrometry identified the 16 kDa Plasmodium falciparum parasitophorous vacuole membrane protein Pfs16 as a potential parasite target. Complementary methods including cellular thermal shift assays confirmed that the parent molecule DDD01035881 stabilised Pfs16 in lysates from activated mature gametocytes. Combined with high-resolution, fluorescence and electron microscopy data, which demonstrated that parasites inhibited with N-4HCS compounds phenocopy the targeted deletion of Pfs16 in gametocytes, these data implicate Pfs16 as a likely target of DDD01035881. This finding establishes N-4HCS compounds as being flexible and effective starting candidates from which transmission-blocking antimalarials can be developed in the future.


Subject(s)
Malaria , Plasmodium , Animals , Male , Membrane Proteins/metabolism , Vacuoles/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Sulfonamides/metabolism
2.
Antimicrob Agents Chemother ; 65(11): e0031121, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34460304

ABSTRACT

Novel bis-1,2,4-triazine compounds with potent in vitro activity against Plasmodium falciparum parasites were recently identified. The bis-1,2,4-triazines represent a unique antimalarial pharmacophore and are proposed to act by a novel but as-yet-unknown mechanism of action. This study investigated the activity of the bis-1,2,4-triazine MIPS-0004373 across the mammalian life cycle stages of the parasite and profiled the kinetics of activity against blood and transmission stage parasites in vitro and in vivo. MIPS-0004373 demonstrated rapid and potent activity against P. falciparum, with excellent in vitro activity against all asexual blood stages. Prolonged in vitro drug exposure failed to generate stable resistance de novo, suggesting a low propensity for the emergence of resistance. Excellent activity was observed against sexually committed ring stage parasites, but activity against mature gametocytes was limited to inhibiting male gametogenesis. Assessment of liver stage activity demonstrated good activity in an in vitro P. berghei model but no activity against Plasmodium cynomolgi hypnozoites or liver schizonts. The bis-1,2,4-triazine MIPS-0004373 efficiently cleared an established P. berghei infection in vivo, with efficacy similar to that of artesunate and chloroquine and a recrudescence profile comparable to that of chloroquine. This study demonstrates the suitability of bis-1,2,4-triazines for further development toward a novel treatment for acute malaria.


Subject(s)
Malaria , Parasites , Animals , Malaria/drug therapy , Male , Plasmodium berghei , Triazines/pharmacology
3.
Article in English | MEDLINE | ID: mdl-33139275

ABSTRACT

Resistance to artemisinin-based combination therapy (ACT) in the Plasmodium falciparum parasite is threatening to reverse recent gains in reducing global deaths from malaria. While resistance manifests as delayed parasite clearance in patients, the phenotype can only spread geographically via the sexual stages and mosquito transmission. In addition to their asexual killing properties, artemisinin and its derivatives sterilize sexual male gametocytes. Whether resistant parasites overcome this sterilizing effect has not, however, been fully tested. Here, we analyzed P. falciparum clinical isolates from the Greater Mekong Subregion, each demonstrating delayed clinical clearance and known resistance-associated polymorphisms in the Kelch13 (PfK13var) gene. As well as demonstrating reduced asexual sensitivity to drug, certain PfK13var isolates demonstrated a marked reduction in sensitivity to artemisinin in an in vitro male gamete formation assay. Importantly, this same reduction in sensitivity was observed when the most resistant isolate was tested directly in mosquito feeds. These results indicate that, under artemisinin drug pressure, while sensitive parasites are blocked, resistant parasites continue transmission. This selective advantage for resistance transmission could favor acquisition of additional host-specificity or polymorphisms affecting partner drug sensitivity in mixed infections. Favored resistance transmission under ACT coverage could have profound implications for the spread of multidrug-resistant malaria beyond Southeast Asia.


Subject(s)
Antimalarials , Artemisinins , Culicidae , Malaria, Falciparum , Parasites , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Asia, Southeastern , Drug Resistance/genetics , Humans , Malaria, Falciparum/drug therapy , Male , Plasmodium falciparum/genetics
4.
J Med Chem ; 63(5): 2240-2262, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31490680

ABSTRACT

Malaria is still a leading cause of mortality among children in the developing world, and despite the immense progress made in reducing the global burden, further efforts are needed if eradication is to be achieved. In this context, targeting transmission is widely recognized as a necessary intervention toward that goal. After carrying out a screen to discover new transmission-blocking agents, herein we report our medicinal chemistry efforts to study the potential of the most robust hit, DDD01035881, as a male-gamete targeted compound. We reveal key structural features for the activity of this series and identify analogues with greater potency and improved metabolic stability. We believe this study lays the groundwork for further development of this series as a transmission blocking agent.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Malaria/transmission , Plasmodium falciparum/drug effects , Animals , Drug Discovery , Female , Germ Cells/drug effects , Hep G2 Cells , Humans , Malaria/drug therapy , Malaria/prevention & control , Male , Mice , Plasmodium falciparum/cytology , Structure-Activity Relationship
5.
Arch Dis Child ; 104(12): 1138-1142, 2019 12.
Article in English | MEDLINE | ID: mdl-31744794

ABSTRACT

OBJECTIVE: The global impact of artemisinin-based combination therapies on malaria-associated mortality and their origins in ancient Chinese medicine has heightened interest in the natural discovery of future antimalarials. METHODS: A double-blind study to identify potential ingredients with antimalarial activity from traditional remedies with reported antipyretic properties. Recipes of clear broths, passed down by tradition in families of diverse ethnic origin, were sourced by school children. Broths were then tested for their ability to arrest malaria parasite asexual growth or sexual stage development in vitro. Clear broth extract was incubated with in vitro cultures of Plasmodium falciparum asexual or mature sexual stage cultures and assayed for parasite viability after 72 hours. RESULTS: Of the 56 broths tested, 5 were found to give >50% in vitro growth inhibition against P. falciparum asexual blood stages, with 2 having comparable inhibition to that seen with dihydroartemisinin, a leading antimalarial. Four other broths were found to have >50% transmission blocking activity, preventing male parasite sexual stage development. After unblinding, two active broths were found to be from siblings from different classes, who had brought in the same vegetarian soup, demonstrating assay robustness. CONCLUSIONS: This screening approach succeeded in finding broths with activity against malaria parasite in vitro growth, arising from complex vegetable and/or meat-based broths. This represented a successful child education exercise, in teaching about the interface between natural remedies, traditional medicine and evidence-based drug discovery.


Subject(s)
Antipyretics/pharmacology , Artemisinins/pharmacology , Drugs, Chinese Herbal/pharmacology , Malaria, Falciparum/diet therapy , Medicine, Chinese Traditional , Plasmodium falciparum/drug effects , Child , Double-Blind Method , Food , Humans , Malaria, Falciparum/prevention & control , Meat , Vegetables
6.
Proc Natl Acad Sci U S A ; 116(14): 7015-7020, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30894487

ABSTRACT

Malaria and cryptosporidiosis, caused by apicomplexan parasites, remain major drivers of global child mortality. New drugs for the treatment of malaria and cryptosporidiosis, in particular, are of high priority; however, there are few chemically validated targets. The natural product cladosporin is active against blood- and liver-stage Plasmodium falciparum and Cryptosporidium parvum in cell-culture studies. Target deconvolution in P. falciparum has shown that cladosporin inhibits lysyl-tRNA synthetase (PfKRS1). Here, we report the identification of a series of selective inhibitors of apicomplexan KRSs. Following a biochemical screen, a small-molecule hit was identified and then optimized by using a structure-based approach, supported by structures of both PfKRS1 and C. parvum KRS (CpKRS). In vivo proof of concept was established in an SCID mouse model of malaria, after oral administration (ED90 = 1.5 mg/kg, once a day for 4 d). Furthermore, we successfully identified an opportunity for pathogen hopping based on the structural homology between PfKRS1 and CpKRS. This series of compounds inhibit CpKRS and C. parvum and Cryptosporidium hominis in culture, and our lead compound shows oral efficacy in two cryptosporidiosis mouse models. X-ray crystallography and molecular dynamics simulations have provided a model to rationalize the selectivity of our compounds for PfKRS1 and CpKRS vs. (human) HsKRS. Our work validates apicomplexan KRSs as promising targets for the development of drugs for malaria and cryptosporidiosis.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum/enzymology , Enzyme Inhibitors/pharmacology , Lysine-tRNA Ligase/antagonists & inhibitors , Malaria, Falciparum , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Animals , Cryptosporidiosis/drug therapy , Cryptosporidiosis/enzymology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Humans , Lysine-tRNA Ligase/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/enzymology , Mice, SCID , Protozoan Proteins/metabolism
7.
Nat Commun ; 9(1): 3805, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228275

ABSTRACT

Spread of parasite resistance to artemisinin threatens current frontline antimalarial therapies, highlighting the need for new drugs with alternative modes of action. Since only 0.2-1% of asexual parasites differentiate into sexual, transmission-competent forms, targeting this natural bottleneck provides a tangible route to interrupt disease transmission and mitigate resistance selection. Here we present a high-throughput screen of gametogenesis against a ~70,000 compound diversity library, identifying seventeen drug-like molecules that target transmission. Hit molecules possess varied activity profiles including male-specific, dual acting male-female and dual-asexual-sexual, with one promising N-((4-hydroxychroman-4-yl)methyl)-sulphonamide scaffold found to have sub-micromolar activity in vitro and in vivo efficacy. Development of leads with modes of action focussed on the sexual stages of malaria parasite development provide a previously unexplored base from which future therapeutics can be developed, capable of preventing parasite transmission through the population.


Subject(s)
Antimalarials/analysis , Drug Evaluation, Preclinical , High-Throughput Screening Assays/methods , Malaria/parasitology , Malaria/transmission , Parasites/physiology , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Feeding Behavior , Female , Gametogenesis/drug effects , Hep G2 Cells , Humans , Male , Mice , Parasites/drug effects , Phenotype , Reproducibility of Results , Structure-Activity Relationship
8.
Malar J ; 17(1): 282, 2018 Aug 03.
Article in English | MEDLINE | ID: mdl-30075783

ABSTRACT

BACKGROUND: The study of malaria transmission requires the experimental infection of mosquitoes with Plasmodium gametocytes. In the laboratory, this is achieved using artificial membrane feeding apparatus that simulate body temperature and skin of the host, and so permit mosquito feeding on reconstituted gametocyte-containing blood. Membrane feeders either use electric heating elements or complex glass chambers to warm the infected blood; both of which are expensive to purchase and can only be sourced from a handful of specialized companies. Presented and tested here is a membrane feeder that can be inexpensively printed using 3D-printing technology. RESULTS: Using the Plasmodium falciparum laboratory strain NF54, three independent standard membrane feeding assays (SMFAs) were performed comparing the 3D-printed feeder against a commercial glass feeder. Exflagellation rates did not differ between the two feeders. Furthermore, no statistically significant difference was found in the oocyst load nor oocyst intensity of Anopheles stephensi mosquitoes (mean oocyst range 1.3-6.2 per mosquito; infection prevalence range 41-79%). CONCLUSIONS: Open source provision of the design files of the 3D-printed feeder will facilitate a wider range of laboratories to perform SMFAs in laboratory and field settings, and enable them to freely customize the design to their own requirements.


Subject(s)
Anopheles/parasitology , Entomology/methods , Malaria, Falciparum/transmission , Membranes, Artificial , Mosquito Vectors/parasitology , Parasitology/methods , Printing, Three-Dimensional/economics , Animals , Humans , Plasmodium falciparum/physiology , Printing, Three-Dimensional/instrumentation
9.
Cell Host Microbe ; 20(5): 618-630, 2016 Nov 09.
Article in English | MEDLINE | ID: mdl-27832590

ABSTRACT

Surface-associated TRAP (thrombospondin-related anonymous protein) family proteins are conserved across the phylum of apicomplexan parasites. TRAP proteins are thought to play an integral role in parasite motility and cell invasion by linking the extracellular environment with the parasite submembrane actomyosin motor. Blood stage forms of the malaria parasite Plasmodium express a TRAP family protein called merozoite-TRAP (MTRAP) that has been implicated in erythrocyte invasion. Using MTRAP-deficient mutants of the rodent-infecting P. berghei and human-infecting P. falciparum parasites, we show that MTRAP is dispensable for erythrocyte invasion. Instead, MTRAP is essential for gamete egress from erythrocytes, where it is necessary for the disruption of the gamete-containing parasitophorous vacuole membrane, and thus for parasite transmission to mosquitoes. This indicates that motor-binding TRAP family members function not just in parasite motility and cell invasion but also in membrane disruption and cell egress.


Subject(s)
Erythrocytes/parasitology , Exocytosis , Merozoites/physiology , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Vacuoles/parasitology , Animals , Culicidae , Humans , Membranes/metabolism , Mice
10.
Nat Protoc ; 11(9): 1668-80, 2016 09.
Article in English | MEDLINE | ID: mdl-27560172

ABSTRACT

The prevention of parasite transmission from the human host to the mosquito has been recognized as a vital tool for malaria eradication campaigns. However, transmission-blocking antimalarial drug and/or vaccine discovery and development is currently hampered by the expense and difficulty of producing mature Plasmodium falciparum gametocytes in vitro-the parasite stage responsible for mosquito infection. Current protocols for P. falciparum gametocyte culture usually require complex parasite synchronization and addition of stimulating and/or inhibitory factors, and they may not have demonstrated the essential property of mosquito infectivity. This protocol details all the steps required for reliable P. falciparum gametocyte production and highlights common factors that influence culture success. The protocol can be completed in 15 d, and particular emphasis is placed upon operating a gametocyte culture facility on a continuous cycle. In addition, we show how functionally viable gametocytes can be used to evaluate transmission-blocking drugs both in a field setting and at high throughput (HTP) for drug discovery.


Subject(s)
Cell Culture Techniques/methods , Germ Cells/cytology , Plasmodium falciparum/cytology , Humans
11.
Malar J ; 13: 468, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25439984

ABSTRACT

BACKGROUND: Gametocytes are the Plasmodium life stage that is solely responsible for malaria transmission. Despite their important role in perpetuating malaria, gametocyte differentiation and development is poorly understood. METHODS: To shed light on the biochemical changes that occur during asexual and gametocyte development, metabolic characterization of media from in vitro intra-erythrocytic Plasmodium falciparum cultures was performed throughout gametocyte development by applying 1H nuclear magnetic spectroscopy, and using sham erythrocyte cultures as controls. Spectral differences between parasite and sham cultures were assessed via principal component analyses and partial-least squares analyses, and univariate statistical methods. RESULTS: Clear parasite-associated changes in metabolism were observed throughout the culture period, revealing differences between asexual parasites and gametocyte stages. With culture progression and development of gametocytes, parasitic release of the glycolytic end products lactate, pyruvate, alanine, and glycerol, were found to be dramatically reduced whilst acetate release was greatly increased. Also, uptake of lipid moieties CH(2), CH(3), and CH = CH-CH(2)-CH(2) increased throughout gametocyte development, peaking with maturity. CONCLUSIONS: This study uniquely presents an initial characterization of the metabolic exchange between parasite and culture medium during in vitro P. falciparum gametocyte culture. Results suggest that energy metabolism and lipid utilization between the asexual stages and gametocytes is different. This study provides new insights for gametocyte-specific nutritional requirements to aid future optimization and standardization of in vitro gametocyte cultivation, and highlights areas of novel gametocyte cell biology that deserve to be studied in greater detail and may yield new targets for transmission-blocking drugs.


Subject(s)
Metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Culture Media/chemistry , Magnetic Resonance Spectroscopy , Phenotype
12.
Cell Host Microbe ; 16(1): 128-40, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25011111

ABSTRACT

Reversible protein phosphorylation regulated by kinases and phosphatases controls many cellular processes. Although essential functions for the malaria parasite kinome have been reported, the roles of most protein phosphatases (PPs) during Plasmodium development are unknown. We report a functional analysis of the Plasmodium berghei protein phosphatome, which exhibits high conservation with the P. falciparum phosphatome and comprises 30 predicted PPs with differential and distinct expression patterns during various stages of the life cycle. Gene disruption analysis of P. berghei PPs reveals that half of the genes are likely essential for asexual blood stage development, whereas six are required for sexual development/sporogony in mosquitoes. Phenotypic screening coupled with transcriptome sequencing unveiled morphological changes and altered gene expression in deletion mutants of two N-myristoylated PPs. These findings provide systematic functional analyses of PPs in Plasmodium, identify how phosphatases regulate parasite development and differentiation, and can inform the identification of drug targets for malaria.


Subject(s)
Gene Expression Regulation , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Plasmodium berghei/enzymology , Plasmodium berghei/growth & development , Animals , Female , Gene Knockout Techniques , Mice , Plasmodium falciparum/enzymology
13.
Cell Microbiol ; 16(5): 657-72, 2014 May.
Article in English | MEDLINE | ID: mdl-23937520

ABSTRACT

Apicomplexan parasites invade host cells by a conserved mechanism: parasite proteins are secreted from apical organelles, anchored in the host cell plasma membrane, and then interact with integral membrane proteins on the zoite surface to form the moving junction (MJ). The junction moves from the anterior to the posterior of the parasite resulting in parasite internalization into the host cell within a parasitophorous vacuole (PV). Conserved as well as coccidia-unique rhoptry neck proteins (RONs) have been described, some of which associate with the MJ. Here we report a novel RON, which we call RON12. RON12 is found only in Plasmodium and is highly conserved across the genus. RON12 lacks a membrane anchor and is a major soluble component of the nascent PV. The bulk of RON12 secretion happens late during invasion (after parasite internalization) allowing accumulation in the fully formed PV with a small proportion of RON12 also apparent occasionally in structures resembling the MJ. RON12, unlike most other RONs is not essential, but deletion of the gene does affect parasite proliferation. The data suggest that although the overall mechanism of invasion by Apicomplexan parasites is conserved, additional components depending on the parasite-host cell combination are required.


Subject(s)
Plasmodium falciparum/growth & development , Protozoan Proteins/metabolism , Endocytosis , Gene Deletion , Plasmodium falciparum/genetics , Protozoan Proteins/genetics
14.
Antimicrob Agents Chemother ; 57(7): 3268-74, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23629698

ABSTRACT

It is the mature gametocytes of Plasmodium that are solely responsible for parasite transmission from the mammalian host to the mosquito. They are therefore a logical target for transmission-blocking antimalarial interventions, which aim to break the cycle of reinfection and reduce the prevalence of malaria cases. Gametocytes, however, are not a homogeneous cell population. They are sexually dimorphic, and both males and females are required for parasite transmission. Using two bioassays, we explored the effects of 20 antimalarials on the functional viability of both male and female mature gametocytes of Plasmodium falciparum. We show that mature male gametocytes (as reported by their ability to produce male gametes, i.e., to exflagellate) are sensitive to antifolates, some endoperoxides, methylene blue, and thiostrepton, with submicromolar 50% inhibitory concentrations (IC50s), whereas female gametocytes (as reported by their ability to activate and form gametes expressing the marker Pfs25) are much less sensitive to antimalarial intervention, with only methylene blue and thiostrepton showing any significant activity. These findings show firstly that the antimalarial responses of male and female gametocytes differ and secondly that the mature male gametocyte should be considered a more vulnerable target than the female gametocyte for transmission-blocking drugs. Given the female-biased sex ratio of Plasmodium falciparum (∼3 to 5 females:1 male), current gametocyte assays without a sex-specific readout are unlikely to identify male-targeted compounds and prioritize them for further development. Both assays reported here are being scaled up to at least medium throughput and will permit identification of key transmission-blocking molecules that have been overlooked by other screening campaigns.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Folic Acid Antagonists/pharmacology , Malaria, Falciparum/drug therapy , Methylene Blue/pharmacology , Plasmodium falciparum/physiology , Thiostrepton/pharmacology
15.
PLoS Pathog ; 8(9): e1002948, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028336

ABSTRACT

Protein phosphorylation and dephosphorylation (catalysed by kinases and phosphatases, respectively) are post-translational modifications that play key roles in many eukaryotic signalling pathways, and are often deregulated in a number of pathological conditions in humans. In the malaria parasite Plasmodium, functional insights into its kinome have only recently been achieved, with over half being essential for blood stage development and another 14 kinases being essential for sexual development and mosquito transmission. However, functions for any of the plasmodial protein phosphatases are unknown. Here, we use reverse genetics in the rodent malaria model, Plasmodium berghei, to examine the role of a unique protein phosphatase containing kelch-like domains (termed PPKL) from a family related to Arabidopsis BSU1. Phylogenetic analysis confirmed that the family of BSU1-like proteins including PPKL is encoded in the genomes of land plants, green algae and alveolates, but not in other eukaryotic lineages. Furthermore, PPKL was observed in a distinct family, separate to the most closely-related phosphatase family, PP1. In our genetic approach, C-terminal GFP fusion with PPKL showed an active protein phosphatase preferentially expressed in female gametocytes and ookinetes. Deletion of the endogenous ppkl gene caused abnormal ookinete development and differentiation, and dissociated apical microtubules from the inner-membrane complex, generating an immotile phenotype and failure to invade the mosquito mid-gut epithelium. These observations were substantiated by changes in localisation of cytoskeletal tubulin and actin, and the micronemal protein CTRP in the knockout mutant as assessed by indirect immunofluorescence. Finally, increased mRNA expression of dozi, a RNA helicase vital to zygote development was observed in ppkl(-) mutants, with global phosphorylation studies of ookinete differentiation from 1.5-24 h post-fertilisation indicating major changes in the first hours of zygote development. Our work demonstrates a stage-specific essentiality of the unique PPKL enzyme, which modulates parasite differentiation, motility and transmission.


Subject(s)
Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Plasmodium berghei/enzymology , Plasmodium berghei/growth & development , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Alveolata/chemistry , Alveolata/genetics , Amino Acid Motifs , Animals , Anopheles/parasitology , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Base Sequence , Cell Differentiation , Genes, Protozoan , Malaria/parasitology , Mice , Mice, Inbred C57BL , Phosphoprotein Phosphatases/genetics , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Protozoan Proteins/genetics , Sequence Analysis, DNA , Viridiplantae/chemistry
16.
PLoS Pathog ; 8(2): e1002554, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22383885

ABSTRACT

Cell-cycle progression is governed by a series of essential regulatory proteins. Two major regulators are cell-division cycle protein 20 (CDC20) and its homologue, CDC20 homologue 1 (CDH1), which activate the anaphase-promoting complex/cyclosome (APC/C) in mitosis, and facilitate degradation of mitotic APC/C substrates. The malaria parasite, Plasmodium, is a haploid organism which, during its life-cycle undergoes two stages of mitosis; one associated with asexual multiplication and the other with male gametogenesis. Cell-cycle regulation and DNA replication in Plasmodium was recently shown to be dependent on the activity of a number of protein kinases. However, the function of cell division cycle proteins that are also involved in this process, such as CDC20 and CDH1 is totally unknown. Here we examine the role of a putative CDC20/CDH1 in the rodent malaria Plasmodium berghei (Pb) using reverse genetics. Phylogenetic analysis identified a single putative Plasmodium CDC20/CDH1 homologue (termed CDC20 for simplicity) suggesting that Plasmodium APC/C has only one regulator. In our genetic approach to delete the endogenous cdc20 gene of P. berghei, we demonstrate that PbCDC20 plays a vital role in male gametogenesis, but is not essential for mitosis in the asexual blood stage. Furthermore, qRT-PCR analysis in parasite lines with deletions of two kinase genes involved in male sexual development (map2 and cdpk4), showed a significant increase in cdc20 transcription in activated gametocytes. DNA replication and ultra structural analyses of cdc20 and map2 mutants showed similar blockage of nuclear division at the nuclear spindle/kinetochore stage. CDC20 was phosphorylated in asexual and sexual stages, but the level of modification was higher in activated gametocytes and ookinetes. Changes in global protein phosphorylation patterns in the Δcdc20 mutant parasites were largely different from those observed in the Δmap2 mutant. This suggests that CDC20 and MAP2 are both likely to play independent but vital roles in male gametogenesis.


Subject(s)
Cell Cycle Proteins/genetics , Gametogenesis/genetics , Plasmodium malariae/genetics , Saccharomyces cerevisiae Proteins/genetics , Amino Acid Sequence , Animals , Cdc20 Proteins , Cdh1 Proteins , Genes, Protozoan/physiology , Germ Cells/metabolism , Germ Cells/physiology , Kinetochores/metabolism , Kinetochores/physiology , Malaria/parasitology , Male , Mice , Molecular Sequence Data , Organisms, Genetically Modified , Phylogeny , Plasmodium malariae/growth & development , Plasmodium malariae/metabolism , Plasmodium malariae/physiology , Sequence Homology
17.
Antimicrob Agents Chemother ; 55(6): 2824-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21402842

ABSTRACT

During blood infection, malarial parasites use D-glucose as their main energy source. The Plasmodium falciparum hexose transporter (PfHT), which mediates the uptake of D-glucose into parasites, is essential for survival of asexual blood-stage parasites. Recently, genetic studies in the rodent malaria model, Plasmodium berghei, found that the orthologous hexose transporter (PbHT) is expressed throughout the parasite's development within the mosquito vector, in addition to being essential during intraerythrocytic development. Here, using a D-glucose-derived specific inhibitor of plasmodial hexose transporters, compound 3361, we have investigated the importance of D-glucose uptake during liver and transmission stages of P. berghei. Initially, we confirmed the expression of PbHT during liver stage development, using a green fluorescent protein (GFP) tagging strategy. Compound 3361 inhibited liver-stage parasite development, with a 50% inhibitory concentration (IC50) of 11 µM. This process was insensitive to the external D-glucose concentration. In addition, compound 3361 inhibited ookinete development and microgametogenesis, with IC50s in the region of 250 µM (the latter in a D-glucose-sensitive manner). Consistent with our findings for the effect of compound 3361 on vector parasite stages, 1 mM compound 3361 demonstrated transmission blocking activity. These data indicate that novel chemotherapeutic interventions that target PfHT may be active against liver and, to a lesser extent, transmission stages, in addition to blood stages.


Subject(s)
Antimalarials/pharmacology , Monosaccharide Transport Proteins/antagonists & inhibitors , Plasmodium berghei/drug effects , Animals , Cell Line, Tumor , Erythrocytes/parasitology , Glucose/pharmacology , Humans , Liver/parasitology , Mice , Plasmodium berghei/growth & development
18.
Clin Sci (Lond) ; 120(12): 515-24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21208193

ABSTRACT

The regulation of human Th17 cell effector function by Treg cells (regulatory T-cells) is poorly understood. In the present study, we report that human Treg (CD4(+)CD25(+)) cells inhibit the proliferative response of Th17 cells but not their capacity to secrete IL (interleukin)-17. However, they could inhibit proliferation and cytokine production by Th1 and Th2 cells as determined by IFN-γ (interferon-γ) and IL-5 biosynthesis. Currently, as there is interest in the role of IL-17-producing cells and Treg cells in chronic inflammatory diseases in humans, we investigated the presence of CD4(+)CD25(+) T-cells and IL-17 in inflammation in the human lung. Transcripts for IL-17 were expressed in mononuclear cells and purified T-cells from lung tissue of patients with chronic pulmonary inflammation and, when activated, these cells secrete soluble protein. The T-cell-specific transcription factors RORCv2 (retinoic acid-related orphan receptor Cv2; for Th17) and FOXP3 (forkhead box P3; for Treg cells) were enriched in the T-cell fraction of lung mononuclear cells. Retrospective stratification of the patient cohort into those with COPD (chronic obstructive pulmonary disease) and non-COPD lung disease revealed no difference in the expression of IL-17 and IL-23 receptor between the groups. We observed that CD4(+)CD25(+) T-cells were present in comparable numbers in COPD and non-COPD lung tissue and with no correlation between the presence of CD4(+)CD25(+) T-cells and IL-17-producing cells. These results suggest that IL-17-expressing cells are present in chronically inflamed lung tissue, but there is no evidence to support this is due to the recruitment or expansion of Treg cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Pneumonia/immunology , Pneumonia/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , Child , Chronic Disease , Female , Humans , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Middle Aged , T-Lymphocytes, Regulatory/metabolism
19.
PLoS One ; 5(9): e12901, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20886115

ABSTRACT

Malaria, caused by the apicomplexan parasite Plasmodium, threatens 40% of the world's population. Transmission between vertebrate and insect hosts depends on the sexual stages of the life-cycle. The male gamete of Plasmodium parasite is the only developmental stage that possesses a flagellum. Very little is known about the identity or function of proteins in the parasite's flagellar biology. Here, we characterise a Plasmodium PF16 homologue using reverse genetics in the mouse malaria parasite Plasmodium berghei. PF16 is a conserved Armadillo-repeat protein that regulates flagellar structure and motility in organisms as diverse as green algae and mice. We show that P. berghei PF16 is expressed in the male gamete flagellum, where it plays a crucial role maintaining the correct microtubule structure in the central apparatus of the axoneme as studied by electron microscopy. Disruption of the PF16 gene results in abnormal flagellar movement and reduced fertility, but does not lead to complete sterility, unlike pf16 mutations in other organisms. Using homology modelling, bioinformatics analysis and complementation studies in Chlamydomonas, we show that some regions of the PF16 protein are highly conserved across all eukaryotes, whereas other regions may have species-specific functions. PF16 is the first ARM-repeat protein characterised in the malaria parasite genus Plasmodium and this study opens up a novel model for analysis of Plasmodium flagellar biology that may provide unique insights into an ancient organelle and suggest novel intervention strategies to control the malaria parasite.


Subject(s)
Flagella/chemistry , Flagella/metabolism , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/metabolism , Plasmodium berghei/growth & development , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Spermatozoa/metabolism , Animals , Armadillo Domain Proteins , Fertility , Flagella/genetics , High Mobility Group Proteins/genetics , Humans , Malaria/parasitology , Male , Mice , Molecular Sequence Data , Plasmodium berghei/chemistry , Plasmodium berghei/genetics , Plasmodium berghei/physiology , Protozoan Proteins/genetics , Spermatozoa/chemistry
20.
Cell Host Microbe ; 8(4): 377-87, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20951971

ABSTRACT

Although eukaryotic protein kinases (ePKs) contribute to many cellular processes, only three Plasmodium falciparum ePKs have thus far been identified as essential for parasite asexual blood stage development. To identify pathways essential for parasite transmission between their mammalian host and mosquito vector, we undertook a systematic functional analysis of ePKs in the genetically tractable rodent parasite Plasmodium berghei. Modeling domain signatures of conventional ePKs identified 66 putative Plasmodium ePKs. Kinomes are highly conserved between Plasmodium species. Using reverse genetics, we show that 23 ePKs are redundant for asexual erythrocytic parasite development in mice. Phenotyping mutants at four life cycle stages in Anopheles stephensi mosquitoes revealed functional clusters of kinases required for sexual development and sporogony. Roles for a putative SR protein kinase (SRPK) in microgamete formation, a conserved regulator of clathrin uncoating (GAK) in ookinete formation, and a likely regulator of energy metabolism (SNF1/KIN) in sporozoite development were identified.


Subject(s)
Anopheles/parasitology , Plasmodium berghei/enzymology , Protein Kinases/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Energy Metabolism , Female , Gene Deletion , Gene Knockout Techniques , Insect Vectors/parasitology , Life Cycle Stages , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Plasmodium berghei/genetics , Plasmodium berghei/physiology , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Sporozoites/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL