Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Immunol ; 212(9): 1467-1478, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38477614

ABSTRACT

Development of Plasmodium-specific humoral immunity is critically dependent on CD4 Th cell responses and germinal center (GC) reactions during blood-stage Plasmodium infection. IL-21, a cytokine primarily produced by CD4 T cells, is an essential regulator of affinity maturation, isotype class-switching, B cell differentiation, and maintenance of GC reactions in response to many infection and immunization models. In models of experimental malaria, mice deficient in IL-21 or its receptor IL-21R fail to develop memory B cell populations and are not protected against secondary infection. However, whether sustained IL-21 signaling in ongoing GCs is required for maintaining GC magnitude, organization, and output is unclear. In this study, we report that CD4+ Th cells maintain IL-21 expression after resolution of primary Plasmodium yoelii infection. We generated an inducible knockout mouse model that enabled cell type-specific and timed deletion of IL-21 in peripheral, mature CD4 T cells. We found that persistence of IL-21 signaling in active GCs had no impact on the magnitude of GC reactions or their capacity to produce memory B cell populations. However, the memory B cells generated in the absence of IL-21 exhibited reduced recall function upon challenge. Our data support that IL-21 prevents premature cellular dissolution within the GC and promotes stringency of selective pressures during B cell fate determination required to produce high-quality Plasmodium-specific memory B cells. These data are additionally consistent with a temporal requirement for IL-21 in fine-tuning humoral immune memory responses during experimental malaria.


Subject(s)
CD4-Positive T-Lymphocytes , Interleukins , Malaria , Plasmodium , Animals , Mice , B-Lymphocytes , CD4-Positive T-Lymphocytes/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Malaria/immunology , Memory B Cells/immunology , Mice, Inbred C57BL , Plasmodium/immunology
2.
Mol Microbiol ; 121(3): 431-452, 2024 03.
Article in English | MEDLINE | ID: mdl-37492994

ABSTRACT

Transmission of malaria parasites to the mosquito is mediated by sexual precursor cells, the gametocytes. Upon entering the mosquito midgut, the gametocytes egress from the enveloping erythrocyte while passing through gametogenesis. Egress follows an inside-out mode during which the membrane of the parasitophorous vacuole (PV) ruptures prior to the erythrocyte membrane. Membrane rupture requires exocytosis of specialized egress vesicles of the parasites; that is, osmiophilic bodies (OBs) involved in rupturing the PV membrane, and vesicles that harbor the perforin-like protein PPLP2 (here termed P-EVs) required for erythrocyte lysis. While some OB proteins have been identified, like G377 and MDV1/Peg3, the majority of egress vesicle-resident proteins is yet unknown. Here, we used high-resolution imaging and BioID methods to study the two egress vesicle types in Plasmodium falciparum gametocytes. We show that OB exocytosis precedes discharge of the P-EVs and that exocytosis of the P-EVs, but not of the OBs, is calcium sensitive. Both vesicle types exhibit distinct proteomes with the majority of proteins located in the OBs. In addition to known egress-related proteins, we identified novel components of OBs and P-EVs, including vesicle-trafficking proteins. Our data provide insight into the immense molecular machinery required for the inside-out egress of P. falciparum gametocytes.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Animals , Plasmodium falciparum/metabolism , Proteomics/methods , Protozoan Proteins/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/parasitology
3.
Proc Natl Acad Sci U S A ; 119(34): e2204167119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35972967

ABSTRACT

Malaria remains a global driver of morbidity and mortality. To generate new antimalarials, one must elucidate the fundamental cell biology of Plasmodium falciparum, the parasite responsible for the deadliest cases of malaria. A membranous and proteinaceous scaffold called the inner membrane complex (IMC) supports the parasite during morphological changes, including segmentation of daughter cells during asexual replication and formation of transmission-stage gametocytes. The basal complex lines the edge of the IMC during segmentation and likely facilitates IMC expansion. It is unknown, however, what drives IMC expansion during gametocytogenesis. We describe the discovery of a basal complex protein, PfBLEB, which we find to be essential for gametocytogenesis. Parasites lacking PfBLEB harbor defects in IMC expansion and are unable to form mature gametocytes. This article demonstrates a role for a basal complex protein outside of asexual division, and, importantly, highlights a potential molecular target for the ablation of malaria transmission.


Subject(s)
Gametogenesis , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Animals , Antimalarials/chemistry , Drug Design , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
4.
J Biol Chem ; 299(7): 104871, 2023 07.
Article in English | MEDLINE | ID: mdl-37247760

ABSTRACT

Malaria causes >600 thousand fatalities each year, with most cases attributed to the human-infectious Plasmodium falciparum species. Many rodent-infectious Plasmodium species, like Plasmodium berghei and Plasmodium yoelii, have been used as model species that can expedite studies of this pathogen. P. yoelii is an especially good model for investigating the mosquito and liver stages of development because key attributes closely resemble those of P. falciparum. Because of its importance, in 2002 the 17XNL strain of P. yoelii was the first rodent malaria parasite to be sequenced. Although this was a breakthrough effort, the assembly consisted of >5000 contiguous sequences that adversely impacted the annotated gene models. While other rodent malaria parasite genomes have been sequenced and annotated since then, including the related P. yoelii 17X strain, the 17XNL strain has not. As a result, genomic data for 17X has become the de facto reference genome for the 17XNL strain while leaving open questions surrounding possible differences between the 17XNL and 17X genomes. In this work, we present a high-quality genome assembly for P. yoelii 17XNL using PacBio DNA sequencing. In addition, we use Nanopore and Illumina RNA sequencing of mixed blood stages to create complete gene models that include coding sequences, alternate isoforms, and UTR designations. A comparison of the 17X and this new 17XNL assembly revealed biologically meaningful differences between the strains due to the presence of coding sequence variants. Taken together, our work provides a new genomic framework for studies with this commonly used rodent malaria model species.


Subject(s)
Malaria , Parasites , Plasmodium yoelii , Animals , Humans , Plasmodium yoelii/genetics , Rodentia , Malaria/parasitology , Liver
5.
PLoS Biol ; 19(10): e3001434, 2021 10.
Article in English | MEDLINE | ID: mdl-34673764

ABSTRACT

Productive transmission of malaria parasites hinges upon the execution of key transcriptional and posttranscriptional regulatory events. While much is now known about how specific transcription factors activate or repress sexual commitment programs, far less is known about the production of a preferred mRNA homeostasis following commitment and through the host-to-vector transmission event. Here, we show that in Plasmodium parasites, the NOT1 scaffold protein of the CAF1/CCR4/Not complex is duplicated, and one paralogue is dedicated for essential transmission functions. Moreover, this NOT1-G paralogue is central to the sex-specific functions previously associated with its interacting partners, as deletion of not1-g in Plasmodium yoelii leads to a comparable or complete arrest phenotype for both male and female parasites. We show that, consistent with its role in other eukaryotes, PyNOT1-G localizes to cytosolic puncta throughout much of the Plasmodium life cycle. PyNOT1-G is essential to both the complete maturation of male gametes and to the continued development of the fertilized zygote originating from female parasites. Comparative transcriptomics of wild-type and pynot1-g- parasites shows that loss of PyNOT1-G leads to transcript dysregulation preceding and during gametocytogenesis and shows that PyNOT1-G acts to preserve mRNAs that are critical to sexual and early mosquito stage development. Finally, we demonstrate that the tristetraprolin (TTP)-binding domain, which acts as the typical organization platform for RNA decay (TTP) and RNA preservation (ELAV/HuR) factors is dispensable for PyNOT1-G's essential blood stage functions but impacts host-to-vector transmission. Together, we conclude that a NOT1-G paralogue in Plasmodium fulfills the complex transmission requirements of both male and female parasites.


Subject(s)
Life Cycle Stages , Parasites/growth & development , Parasites/metabolism , Plasmodium/growth & development , Plasmodium/metabolism , Protozoan Proteins/metabolism , Sequence Homology, Amino Acid , Animals , Cytosol/metabolism , Female , Gene Duplication , Gene Expression Regulation, Developmental , Germ Cells/physiology , Male , Mice , Models, Biological , Protein Domains , Protein Interaction Maps , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Sexual Maturation/physiology , Transcriptome/genetics , Zygote/growth & development
6.
PLoS Pathog ; 15(1): e1007164, 2019 01.
Article in English | MEDLINE | ID: mdl-30703164

ABSTRACT

With relatively few known specific transcription factors to control the abundance of specific mRNAs, Plasmodium parasites may rely more on the regulation of transcript stability and turnover to provide sufficient gene regulation. Plasmodium transmission stages impose translational repression on specific transcripts in part to accomplish this. However, few proteins are known to participate in this process, and those that are characterized primarily affect female gametocytes. We have identified and characterized Plasmodium yoelii (Py) CCR4-1, a putative deadenylase, which plays a role in the development and activation of male gametocytes, regulates the abundance of specific mRNAs in gametocytes, and ultimately increases the efficiency of host-to-vector transmission. We find that when pyccr4-1 is deleted or its protein made catalytically inactive, there is a loss in the initial coordination of male gametocyte maturation and a reduction of parasite infectivity of the mosquito. Expression of only the N-terminal CAF1 domain of the essential CAF1 deadenylase leads to a similar phenotype. Comparative RNA-seq revealed that PyCCR4-1 affects transcripts important for transmission-related functions that are associated with male or female gametocytes, some of which directly associate with the immunoprecipitated complex. Finally, circular RT-PCR of one of the bound, dysregulated transcripts showed that deletion of the pyccr4-1 gene does not result in gross changes to its UTR or poly(A) tail length. We conclude that the two putative deadenylases of the CAF1/CCR4/NOT complex play critical and intertwined roles in gametocyte maturation and transmission.


Subject(s)
Plasmodium falciparum/genetics , Receptors, CCR4/metabolism , Animals , Culicidae/metabolism , Exoribonucleases , Gametogenesis/physiology , Gene Expression Regulation , Homeodomain Proteins , Male , Mice , Mosquito Vectors , Plasmodium/genetics , Plasmodium falciparum/metabolism , Proteins , RNA, Messenger/genetics , Repressor Proteins , Ribonucleases , Transcription Factors/metabolism , Transcriptional Activation
7.
J Biol Chem ; 294(24): 9555-9566, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31043479

ABSTRACT

Malaria remains a major global health issue, affecting millions and killing hundreds of thousands of people annually. Efforts to break the transmission cycle of the causal Plasmodium parasite, and to cure those that are afflicted, rely upon functional characterization of genes essential to the parasite's growth and development. These studies are often based upon manipulations of the parasite genome to disrupt or modify a gene of interest to understand its importance and function. However, these approaches can be limited by the availability of selectable markers and the time required to generate transgenic parasites. Moreover, there also is a risk of disrupting native gene regulatory elements with the introduction of exogenous sequences. To address these limitations, we have developed CRISPR-RGR, a Streptococcus pyogenes (Sp)Cas9-based gene editing system for Plasmodium that utilizes a ribozyme-guide-ribozyme (RGR) single guide RNA (sgRNA) expression strategy with RNA polymerase II promoters. Using rodent-infectious Plasmodium yoelii, we demonstrate that both gene disruptions and coding sequence insertions are efficiently generated, producing marker-free parasites with homology arms as short as 80-100 bp. Additionally, we find that the common practice of using one sgRNA can produce both unintended plasmid integration and desired locus replacement editing events, whereas the use of two sgRNAs results in only locus replacement editing. Lastly, we show that CRISPR-RGR can be used for CRISPR interference (CRISPRi) by binding catalytically dead SpCas9 (dSpCas9) to the region upstream of a gene of interest, resulting in a position-dependent, but strand-independent reduction in gene expression. This robust and flexible system facilitates efficient genetic characterizations of rodent-infectious Plasmodium species.


Subject(s)
CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Deletion , Gene Editing/methods , Malaria/veterinary , RNA, Catalytic , Rodent Diseases/parasitology , Animals , CRISPR-Associated Protein 9/genetics , Female , Genetic Vectors , Malaria/parasitology , Mice , Plasmids , Plasmodium yoelii/genetics , Plasmodium yoelii/isolation & purification
8.
J Cell Sci ; 131(6)2018 03 26.
Article in English | MEDLINE | ID: mdl-29487181

ABSTRACT

In this study, we characterized the Puf family gene member Puf3 in the malaria parasites Plasmodium falciparum and Plasmodium yoelii Secondary structure prediction suggested that the RNA-binding domains of the Puf3 proteins consisted of 11 pumilio repeats that were similar to those in the human Puf-A (also known as PUM3) and Saccharomyces cerevisiae Puf6 proteins, which are involved in ribosome biogenesis. Neither P. falciparum (Pf)Puf3 nor P. yoelii (Py)Puf3 could be genetically disrupted, suggesting they may be essential for the intraerythrocytic developmental cycle. Cellular fractionation of PfPuf3 in the asexual stages revealed preferential partitioning to the nuclear fraction, consistent with nuclear localization of PfPuf3::GFP and PyPuf3::GFP as detected by immunofluorescence. Furthermore, PfPuf3 colocalized with the nucleolar marker PfNop1, demonstrating that PfPuf3 is a nucleolar protein in the asexual stages. We found, however, that PyPuf3 changed its localization from being nucleolar to being present in cytosolic puncta in the mosquito and liver stages, which may reflect alternative functions in these stages. Affinity purification of molecules that associated with a PTP-tagged variant of PfPuf3 revealed 31 proteins associated with the 60S ribosome, and an enrichment of 28S rRNA and internal transcribed spacer 2 sequences. Taken together, these results suggest an essential function for PfPuf3 in ribosomal biogenesis.


Subject(s)
Plasmodium falciparum/metabolism , Plasmodium yoelii/metabolism , Protozoan Proteins/chemistry , Ribosomes/metabolism , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Cytosol/metabolism , Life Cycle Stages , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium yoelii/chemistry , Plasmodium yoelii/genetics , Plasmodium yoelii/growth & development , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomes/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
10.
PLoS Pathog ; 14(8): e1007249, 2018 08.
Article in English | MEDLINE | ID: mdl-30133543

ABSTRACT

The complex life-cycle of the human malaria parasite Plasmodium falciparum requires a high degree of tight coordination allowing the parasite to adapt to changing environments. One of the major challenges for the parasite is the human-to-mosquito transmission, which starts with the differentiation of blood stage parasites into the transmissible gametocytes, followed by the rapid conversion of the gametocytes into gametes, once they are taken up by the blood-feeding Anopheles vector. In order to pre-adapt to this change of host, the gametocytes store transcripts in stress granules that encode proteins needed for parasite development in the mosquito. Here we report on a novel stress granule component, the seven-helix protein 7-Helix-1. The protein, a homolog of the human stress response regulator LanC-like 2, accumulates in stress granules of female gametocytes and interacts with ribonucleoproteins, such as CITH, DOZI, and PABP1. Malaria parasites lacking 7-Helix-1 are significantly impaired in female gametogenesis and thus transmission to the mosquito. Lack of 7-Helix-1 further leads to a deregulation of components required for protein synthesis. Consistently, inhibitors of translation could mimic the 7-Helix-1 loss-of-function phenotype. 7-Helix-1 forms a complex with the RNA-binding protein Puf2, a translational regulator of the female-specific antigen Pfs25, as well as with pfs25-coding mRNA. In accord, gametocytes deficient of 7-Helix-1 exhibit impaired Pfs25 synthesis. Our data demonstrate that 7-Helix-1 constitutes stress granules crucial for regulating the synthesis of proteins needed for life-cycle progression of Plasmodium in the mosquito vector.


Subject(s)
Anopheles/parasitology , Malaria, Falciparum/transmission , Membrane Proteins/physiology , Plasmodium falciparum , Protein Biosynthesis , Animals , Cytoplasmic Granules/metabolism , Female , Humans , Life Cycle Stages/genetics , Malaria, Falciparum/parasitology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Organisms, Genetically Modified , Phosphate-Binding Proteins , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protein Biosynthesis/genetics , Protein Processing, Post-Translational , Protein Structure, Secondary , Protozoan Proteins/metabolism , Protozoan Proteins/physiology , Sequence Homology , Stress, Physiological
11.
Malar J ; 19(1): 424, 2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33228734

ABSTRACT

BACKGROUND: Well-defined promoters are essential elements for genetic studies in all organisms, and enable controlled expression of endogenous genes, transgene expression, and gene editing. Despite this, there is a paucity of defined promoters for the rodent-infectious malaria parasites. This is especially true for Plasmodium yoelii, which is often used to study the mosquito and liver stages of malarial infection, as well as host immune responses to infection. METHODS: Here six promoters were selected from across the parasite's life cycle (clag-a, dynein heavy chain delta, lap4, trap, uis4, lisp2) that have been invoked in the literature as controlling their genes in a stage-specific manner. A minimal promoter length for the constitutive pybip promoter that confers strong expression levels was also determined, which is useful for expression of reporters and gene editing enzymes. RESULTS: Instead, it was observed that these promoters confer stage-enriched gene control, as some parasites also effectively use these promoters in other stages. Thus, when used alone, these promoters could complicate the interpretation of results obtained from promoter swaps, stage-targeted recombination, or gene editing experiments. CONCLUSIONS: Together these data indicate that achieving stage-specific effects, such as gene editing, is likely best done using a two-component system with independent promoter activities overlapping only in the intended life cycle stage.


Subject(s)
Genes, Protozoan , Malaria/physiopathology , Plasmodium yoelii/genetics , Promoter Regions, Genetic , Animals , Female , Mice
12.
Mol Microbiol ; 106(2): 266-284, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28787542

ABSTRACT

Transmission of the malaria parasite occurs in an unpredictable moment, when a mosquito takes a blood meal. Plasmodium has therefore evolved strategies to prepare for transmission, including translationally repressing and protecting mRNAs needed to establish the infection. However, mechanisms underlying these critical controls are not well understood, including whether Plasmodium changes its translationally repressive complexes and mRNA targets in different stages. Efforts to understand this have been stymied by severe technical limitations due to substantial mosquito contamination of samples. Here using P. yoelii, for the first time we provide a proteomic comparison of a protein complex across asexual blood, sexual and sporozoite stages, along with a transcriptomic comparison of the mRNAs that are affected in these stages. We find that the Apicomplexan-specific ALBA4 RNA-binding protein acts to regulate development of the parasite's transmission stages, and that ALBA4 associates with both stage-specific and stage-independent partners to produce opposing mRNA fates. These efforts expand our understanding and ability to interrogate both sexual and sporozoite transmission stages and the molecular preparations they evolved to perpetuate their infectious cycle.


Subject(s)
Plasmodium yoelii/physiology , RNA, Messenger/biosynthesis , Animals , Anopheles/parasitology , Enzyme Repression , Malaria/parasitology , Parasites , Parasitic Diseases/genetics , Plasmodium yoelii/genetics , Plasmodium yoelii/growth & development , Proteomics , Protozoan Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Sporozoites/metabolism , Transcriptome
13.
PLoS Pathog ; 12(4): e1005606, 2016 04.
Article in English | MEDLINE | ID: mdl-27128092

ABSTRACT

Malaria parasite infection is initiated by the mosquito-transmitted sporozoite stage, a highly motile invasive cell that targets hepatocytes in the liver for infection. A promising approach to developing a malaria vaccine is the use of proteins located on the sporozoite surface as antigens to elicit humoral immune responses that prevent the establishment of infection. Very little of the P. falciparum genome has been considered as potential vaccine targets, and candidate vaccines have been almost exclusively based on single antigens, generating the need for novel target identification. The most advanced malaria vaccine to date, RTS,S, a subunit vaccine consisting of a portion of the major surface protein circumsporozoite protein (CSP), conferred limited protection in Phase III trials, falling short of community-established vaccine efficacy goals. In striking contrast to the limited protection seen in current vaccine trials, sterilizing immunity can be achieved by immunization with radiation-attenuated sporozoites, suggesting that more potent protection may be achievable with a multivalent protein vaccine. Here, we provide the most comprehensive analysis to date of proteins located on the surface of or secreted by Plasmodium falciparum salivary gland sporozoites. We used chemical labeling to isolate surface-exposed proteins on sporozoites and identified these proteins by mass spectrometry. We validated several of these targets and also provide evidence that components of the inner membrane complex are in fact surface-exposed and accessible to antibodies in live sporozoites. Finally, our mass spectrometry data provide the first direct evidence that the Plasmodium surface proteins CSP and TRAP are glycosylated in sporozoites, a finding that could impact the selection of vaccine antigens.


Subject(s)
Malaria, Falciparum/metabolism , Proteomics/methods , Protozoan Proteins/analysis , Protozoan Proteins/metabolism , Sporozoites/metabolism , Animals , Culicidae , Fluorescent Antibody Technique , Glycosylation , Mass Spectrometry , Membrane Proteins/analysis , Membrane Proteins/metabolism , Organisms, Genetically Modified , Sporozoites/chemistry
14.
Proteomics ; 16(23): 2967-2976, 2016 12.
Article in English | MEDLINE | ID: mdl-27714937

ABSTRACT

Accurate and comprehensive identification of surface-exposed proteins (SEPs) in parasites is a key step in developing novel subunit vaccines. However, the reliability of MS-based high-throughput methods for proteome-wide mapping of SEPs continues to be limited due to high rates of false positives (i.e., proteins mistakenly identified as surface exposed) as well as false negatives (i.e., SEPs not detected due to low expression or other technical limitations). We propose a framework called PlasmoSEP for the reliable identification of SEPs using a novel semisupervised learning algorithm that combines SEPs identified by high-throughput experiments and expert annotation of high-throughput data to augment labeled data for training a predictive model. Our experiments using high-throughput data from the Plasmodium falciparum surface-exposed proteome provide several novel high-confidence predictions of SEPs in P. falciparum and also confirm expert annotations for several others. Furthermore, PlasmoSEP predicts that 25 of 37 experimentally identified SEPs in Plasmodium yoelii salivary gland sporozoites are likely to be SEPs. Finally, PlasmoSEP predicts several novel SEPs in P. yoelii and Plasmodium vivax malaria parasites that can be validated for further vaccine studies. Our computational framework can be easily adapted to improve the interpretation of data from high-throughput studies.


Subject(s)
Algorithms , Membrane Proteins/analysis , Plasmodium falciparum/chemistry , Proteomics/methods , Protozoan Proteins/analysis , High-Throughput Screening Assays/methods , Humans , Membrane Proteins/metabolism , Models, Theoretical , Plasmodium vivax/metabolism , Plasmodium vivax/pathogenicity , Plasmodium yoelii/chemistry , Protozoan Proteins/metabolism , Salivary Glands/metabolism
16.
Bioorg Med Chem Lett ; 26(22): 5487-5491, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27780638

ABSTRACT

Plasmodium falciparum calcium-dependent protein kinase 4 (PfCDPK4) is essential for the exflagellation of male gametocytes. Inhibition of PfCDPK4 is an effective way of blocking the transmission of malaria by mosquitoes. A series of 5-aminopyrazole-4-carboxamide analogues are demonstrated to be potent inhibitors of PfCDPK4. The compounds are also able to block exflagellation of Plasmodium falciparum male gametocytes without observable toxicity to mammalian cells.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protein Kinases/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology , Animals , Cell Line , Culicidae/parasitology , Humans , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Male , Plasmodium falciparum/physiology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
17.
BMC Genomics ; 16: 890, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26525978

ABSTRACT

BACKGROUND: The malaria parasites in the genus Plasmodium have a very complicated life cycle involving an invertebrate vector and a vertebrate host. RNA-binding proteins (RBPs) are critical factors involved in every aspect of the development of these parasites. However, very few RBPs have been functionally characterized to date in the human parasite Plasmodium falciparum. METHODS: Using different bioinformatic methods and tools we searched P. falciparum genome to list and annotate RBPs. A representative 3D models for each of the RBD domain identified in P. falciparum was created using I-TESSAR and SWISS-MODEL. Microarray and RNAseq data analysis pertaining PfRBPs was performed using MeV software. Finally, Cytoscape was used to create protein-protein interaction network for CITH-Dozi and Caf1-CCR4-Not complexes. RESULTS: We report the identification of 189 putative RBP genes belonging to 13 different families in Plasmodium, which comprise 3.5% of all annotated genes. Almost 90% (169/189) of these genes belong to six prominent RBP classes, namely RNA recognition motifs, DEAD/H-box RNA helicases, K homology, Zinc finger, Puf and Alba gene families. Interestingly, almost all of the identified RNA-binding helicases and KH genes have cognate homologs in model species, suggesting their evolutionary conservation. Exploration of the existing P. falciparum blood-stage transcriptomes revealed that most RBPs have peak mRNA expression levels early during the intraerythrocytic development cycle, which taper off in later stages. Nearly 27% of RBPs have elevated expression in gametocytes, while 47 and 24% have elevated mRNA expression in ookinete and asexual stages. Comparative interactome analyses using human and Plasmodium protein-protein interaction datasets suggest extensive conservation of the PfCITH/PfDOZI and PfCaf1-CCR4-NOT complexes. CONCLUSIONS: The Plasmodium parasites possess a large number of putative RBPs belonging to most of RBP families identified so far, suggesting the presence of extensive post-transcriptional regulation in these parasites. Taken together, in silico identification of these putative RBPs provides a foundation for future functional studies aimed at defining a unique network of post-transcriptional regulation in P. falciparum.


Subject(s)
Computational Biology , Plasmodium/genetics , Protozoan Proteins/genetics , RNA-Binding Proteins/genetics , RNA, Messenger/genetics
18.
Mol Microbiol ; 91(4): 679-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24330260

ABSTRACT

Malaria parasites scavenge nutrients from their host but also harbour enzymatic pathways for de novo macromolecule synthesis. One such pathway is apicoplast-targeted type II fatty acid synthesis, which is essential for late liver-stage development in rodent malaria. It is likely that fatty acids synthesized in the apicoplast are ultimately incorporated into membrane phospholipids necessary for exoerythrocytic merozoite formation. We hypothesized that these synthesized fatty acids are being utilized for apicoplast-targeted phosphatidic acid synthesis, the phospholipid precursor. Phosphatidic acid is typically synthesized in a three-step reaction utilizing three enzymes: glycerol 3-phosphate dehydrogenase, glycerol 3-phosphate acyltransferase and lysophosphatidic acid acyltransferase. The Plasmodium genome is predicted to harbour genes for both apicoplast- and cytosol/endoplasmic reticulum-targeted phosphatidic acid synthesis. Our research shows that apicoplast-targeted Plasmodium yoelii glycerol 3-phosphate dehydrogenase and glycerol 3-phosphate acyltransferase are expressed only during liver-stage development and deletion of the encoding genes resulted in late liver-stage growth arrest and lack of merozoite differentiation. However, the predicted apicoplast-targeted lysophosphatidic acid acyltransferase gene was refractory to deletion and was expressed solely in the endoplasmic reticulum throughout the parasite life cycle. Our results suggest that P. yoelii has an incomplete apicoplast-targeted phosphatidic acid synthesis pathway that is essential for liver-stage maturation.


Subject(s)
Liver/parasitology , Phosphatidic Acids/biosynthesis , Plasmodium yoelii/enzymology , Plasmodium yoelii/physiology , Plastids/enzymology , Plastids/metabolism , Acyltransferases/metabolism , Animals , Glycerol-3-Phosphate Dehydrogenase (NAD+)/metabolism , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Mice , Plasmodium yoelii/metabolism , Protein Transport
19.
Mol Cell Proteomics ; 12(5): 1127-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23325771

ABSTRACT

Malaria infections of mammals are initiated by the transmission of Plasmodium salivary gland sporozoites during an Anopheles mosquito vector bite. Sporozoites make their way through the skin and eventually to the liver, where they infect hepatocytes. Blocking this initial stage of infection is a promising malaria vaccine strategy. Therefore, comprehensively elucidating the protein composition of sporozoites will be invaluable in identifying novel targets for blocking infection. Previous efforts to identify the proteins expressed in Plasmodium mosquito stages were hampered by the technical difficulty of separating the parasite from its vector; without effective purifications, the large majority of proteins identified were of vector origin. Here we describe the proteomic profiling of highly purified salivary gland sporozoites from two Plasmodium species: human-infective Plasmodium falciparum and rodent-infective Plasmodium yoelii. The combination of improved sample purification and high mass accuracy mass spectrometry has facilitated the most complete proteome coverage to date for a pre-erythrocytic stage of the parasite. A total of 1991 P. falciparum sporozoite proteins and 1876 P. yoelii sporozoite proteins were identified, with >86% identified with high sequence coverage. The proteomic data were used to confirm the presence of components of three features critical for sporozoite infection of the mammalian host: the sporozoite motility and invasion apparatus (glideosome), sporozoite signaling pathways, and the contents of the apical secretory organelles. Furthermore, chemical labeling and identification of proteins on live sporozoites revealed previously uncharacterized complexity of the putative sporozoite surface-exposed proteome. Taken together, the data constitute the most comprehensive analysis to date of the protein expression of salivary gland sporozoites and reveal novel potential surface-exposed proteins that might be valuable targets for antibody blockage of infection.


Subject(s)
Anopheles/parasitology , Insect Vectors/parasitology , Proteome/metabolism , Protozoan Proteins/metabolism , Salivary Glands/parasitology , Sporozoites/metabolism , Animals , Calcium Signaling , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Erythrocytes/parasitology , Female , Host-Parasite Interactions , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Mice , Plasmodium falciparum/metabolism , Plasmodium yoelii/metabolism , Proteome/isolation & purification , Proteomics , Protozoan Proteins/isolation & purification , Secretory Pathway
20.
J Biol Chem ; 288(45): 32539-32552, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24072705

ABSTRACT

The malaria parasite Plasmodium falciparum and related organisms possess a relict plastid known as the apicoplast. Apicoplast protein synthesis is a validated drug target in malaria because antibiotics that inhibit translation in prokaryotes also inhibit apicoplast protein synthesis and are sometimes used for malaria prophylaxis or treatment. We identified components of an indirect aminoacylation pathway for Gln-tRNA(Gln) biosynthesis in Plasmodium that we hypothesized would be essential for apicoplast protein synthesis. Here, we report our characterization of the first enzyme in this pathway, the apicoplast glutamyl-tRNA synthetase (GluRS). We expressed the recombinant P. falciparum enzyme in Escherichia coli, showed that it is nondiscriminating because it glutamylates both apicoplast tRNA(Glu) and tRNA(Gln), determined its kinetic parameters, and demonstrated its inhibition by a known bacterial GluRS inhibitor. We also localized the Plasmodium berghei ortholog to the apicoplast in blood stage parasites but could not delete the PbGluRS gene. These data show that Gln-tRNA(Gln) biosynthesis in the Plasmodium apicoplast proceeds via an essential indirect aminoacylation pathway that is reminiscent of bacteria and plastids.


Subject(s)
Apicoplasts/enzymology , Glutamate-tRNA Ligase/metabolism , Plasmodium berghei/enzymology , Plasmodium falciparum/enzymology , Protein Biosynthesis/physiology , Protozoan Proteins/metabolism , Transfer RNA Aminoacylation/physiology , Apicoplasts/genetics , Glutamate-tRNA Ligase/genetics , Humans , Plasmodium berghei/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , RNA, Transfer, Gln/genetics , RNA, Transfer, Gln/metabolism , RNA, Transfer, Glu/genetics , RNA, Transfer, Glu/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL