Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 21(4): e3002066, 2023 04.
Article in English | MEDLINE | ID: mdl-37053271

ABSTRACT

With emerging resistance to frontline treatments, it is vital that new antimalarial drugs are identified to target Plasmodium falciparum. We have recently described a compound, MMV020291, as a specific inhibitor of red blood cell (RBC) invasion, and have generated analogues with improved potency. Here, we generated resistance to MMV020291 and performed whole genome sequencing of 3 MMV020291-resistant populations. This revealed 3 nonsynonymous single nucleotide polymorphisms in 2 genes; 2 in profilin (N154Y, K124N) and a third one in actin-1 (M356L). Using CRISPR-Cas9, we engineered these mutations into wild-type parasites, which rendered them resistant to MMV020291. We demonstrate that MMV020291 reduces actin polymerisation that is required by the merozoite stage parasites to invade RBCs. Additionally, the series inhibits the actin-1-dependent process of apicoplast segregation, leading to a delayed death phenotype. In vitro cosedimentation experiments using recombinant P. falciparum proteins indicate that potent MMV020291 analogues disrupt the formation of filamentous actin in the presence of profilin. Altogether, this study identifies the first compound series interfering with the actin-1/profilin interaction in P. falciparum and paves the way for future antimalarial development against the highly dynamic process of actin polymerisation.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Plasmodium falciparum/metabolism , Actins/genetics , Actins/metabolism , Profilins/genetics , Profilins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Malaria, Falciparum/genetics , Erythrocytes/parasitology , Antimalarials/pharmacology
2.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068393

ABSTRACT

The highly complex life cycle of the human malaria parasite, Plasmodium falciparum, is based on an orchestrated and tightly regulated gene expression program. In general, eukaryotic transcription regulation is determined by a combination of sequence-specific transcription factors binding to regulatory DNA elements and the packaging of DNA into chromatin as an additional layer. The accessibility of regulatory DNA elements is controlled by the nucleosome occupancy and changes of their positions by an active process called nucleosome remodeling. These epigenetic mechanisms are poorly explored in P. falciparum. The parasite genome is characterized by an extraordinarily high AT-content and the distinct architecture of functional elements, and chromatin-related proteins also exhibit high sequence divergence compared to other eukaryotes. Together with the distinct biochemical properties of nucleosomes, these features suggest substantial differences in chromatin-dependent regulation. Here, we highlight the peculiarities of epigenetic mechanisms in P. falciparum, addressing chromatin structure and dynamics with respect to their impact on transcriptional control. We focus on the specialized chromatin remodeling enzymes and discuss their essential function in P. falciparum gene regulation.


Subject(s)
Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Regulation , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Transcription, Genetic , Animals , Humans , Life Cycle Stages
3.
Cell Microbiol ; 23(11): e13345, 2021 11.
Article in English | MEDLINE | ID: mdl-33885206

ABSTRACT

The cytoskeletal protein actin is highly abundant and conserved in eukaryotic cells. It occurs in two different states- the globular (G-actin) form, which can polymerise into the filamentous (F-actin) form, fulfilling various critical functions including cytokinesis, cargo trafficking and cellular motility. In higher eukaryotes, there are several actin isoforms with nearly identical amino acid sequences. Despite the high level of amino acid identity, they display regulated expression patterns and unique non-redundant roles. The number of actin isoforms together with conserved sequences may reflect the selective pressure exerted by scores of actin binding proteins (ABPs) in higher eukaryotes. In contrast, in many protozoans such as apicomplexan parasites which possess only a few ABPs, the regulatory control of actin and its multiple functions are still obscure. Here, we provide a summary of the regulation and biological functions of actin in higher eukaryotes and compare it with the current knowledge in apicomplexans. We discuss future experiments that will help us understand the multiple, critical roles of this fascinating system in apicomplexans.


Subject(s)
Actins , Parasites , Actin Cytoskeleton , Actins/genetics , Animals , Cell Movement , Microfilament Proteins
4.
EMBO Rep ; 20(12): e48896, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31584242

ABSTRACT

The obligate intracellular parasites Toxoplasma gondii and Plasmodium spp. invade host cells by injecting a protein complex into the membrane of the targeted cell that bridges the two cells through the assembly of a ring-like junction. This circular junction stretches while the parasites apply a traction force to pass through, a step that typically concurs with transient constriction of the parasite body. Here we analyse F-actin dynamics during host cell invasion. Super-resolution microscopy and real-time imaging highlighted an F-actin pool at the apex of pre-invading parasite, an F-actin ring at the junction area during invasion but also networks of perinuclear and posteriorly localised F-actin. Mutant parasites with dysfunctional acto-myosin showed significant decrease of junctional and perinuclear F-actin and are coincidently affected in nuclear passage through the junction. We propose that the F-actin machinery eases nuclear passage by stabilising the junction and pushing the nucleus through the constriction. Our analysis suggests that the junction opposes resistance to the passage of the parasite's nucleus and provides the first evidence for a dual contribution of actin-forces during host cell invasion by apicomplexan parasites.


Subject(s)
Actins/physiology , Host-Parasite Interactions/physiology , Plasmodium falciparum/physiology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/physiology , Toxoplasma/parasitology , Toxoplasma/pathogenicity , Actins/genetics , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/parasitology , Cell Nucleus/physiology , Cells, Cultured , Gene Knockout Techniques , Humans , Merozoites/genetics , Merozoites/pathogenicity , Merozoites/physiology , Models, Biological , Mutation , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Signal Transduction , Toxoplasma/genetics , Virulence/physiology
5.
Elife ; 82019 07 19.
Article in English | MEDLINE | ID: mdl-31322501

ABSTRACT

In addition to its role in erythrocyte invasion, Plasmodium falciparum actin is implicated in endocytosis, cytokinesis and inheritance of the chloroplast-like organelle called the apicoplast. Previously, the inability to visualise filamentous actin (F-actin) dynamics had restricted the characterisation of both F-actin and actin regulatory proteins, a limitation we recently overcame for Toxoplasma (Periz et al, 2017). Here, we have expressed and validated actin-binding chromobodies as F-actin-sensors in Plasmodium falciparum and characterised in-vivo actin dynamics. F-actin could be chemically modulated, and genetically disrupted upon conditionally deleting actin-1. In a comparative approach, we demonstrate that Formin-2, a predicted nucleator of F-actin, is responsible for apicoplast inheritance in both Plasmodium and Toxoplasma, and additionally mediates efficient cytokinesis in Plasmodium. Finally, time-averaged local intensity measurements of F-actin in Toxoplasma conditional mutants revealed molecular determinants of spatiotemporally regulated F-actin flow. Together, our data indicate that Formin-2 is the primary F-actin nucleator during apicomplexan intracellular growth, mediating multiple essential functions.


Subject(s)
Actin Cytoskeleton/metabolism , Cytokinesis/genetics , Formins/chemistry , Malaria, Falciparum/genetics , Actin Cytoskeleton/chemistry , Actins/genetics , Actins/metabolism , Apicoplasts/chemistry , Apicoplasts/metabolism , Endocytosis/genetics , Erythrocytes/chemistry , Erythrocytes/parasitology , Formins/genetics , Gene Expression Regulation/genetics , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/metabolism , Protein Binding , Toxoplasma/metabolism , Toxoplasma/pathogenicity
6.
BMC Biol ; 15(1): 70, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28810863

ABSTRACT

BACKGROUND: The phylum Apicomplexa includes intracellular parasites causing immense global disease burden, the deadliest of them being the human malaria parasite Plasmodium falciparum, which invades and replicates within erythrocytes. The cytoskeletal protein actin is well conserved within apicomplexans but divergent from mammalian actins, and was primarily reported to function during host cell invasion. However, novel invasion mechanisms have been described for several apicomplexans, and specific functions of the acto-myosin system are being reinvestigated. Of the two actin genes in P. falciparum, actin-1 (pfact1) is ubiquitously expressed in all life-cycle stages and is thought to be required for erythrocyte invasion, although its functions during parasite development are unknown, and definitive in vivo characterisation during invasion is lacking. RESULTS: Here we have used a conditional Cre-lox system to investigate the functions of PfACT1 during P. falciparum blood-stage development and host cell invasion. We demonstrate that PfACT1 is crucially required for segregation of the plastid-like organelle, the apicoplast, and for efficient daughter cell separation during the final stages of cytokinesis. Surprisingly, we observe that egress from the host cell is not an actin-dependent process. Finally, we show that parasites lacking PfACT1 are capable of microneme secretion, attachment and formation of a junction with the erythrocyte, but are incapable of host cell invasion. CONCLUSIONS: This study provides important mechanistic insights into the definitive essential functions of PfACT1 in P. falciparum, which are not only of biological interest, but owing to functional divergence from mammalian actins, could also form the basis for the development of novel therapeutics against apicomplexans.


Subject(s)
Actins/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Actins/metabolism , Erythrocytes/parasitology , Host-Parasite Interactions , Humans , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism
7.
PLoS One ; 11(6): e0157873, 2016.
Article in English | MEDLINE | ID: mdl-27332706

ABSTRACT

Malaria is caused by an obligate intracellular protozoan parasite that replicates within and destroys erythrocytes. Asexual blood stages of the causative agent of the most virulent form of human malaria, Plasmodium falciparum, can be cultivated indefinitely in vitro in human erythrocytes, facilitating experimental analysis of parasite cell biology, biochemistry and genetics. However, efforts to improve understanding of the basic biology of this important pathogen and to develop urgently required new antimalarial drugs and vaccines, suffer from a paucity of basic research tools. This includes a simple means of quantifying the effects of drugs, antibodies and gene modifications on parasite fitness and replication rates. Here we describe the development and validation of an extremely simple, robust plaque assay that can be used to visualise parasite replication and resulting host erythrocyte destruction at the level of clonal parasite populations. We demonstrate applications of the plaque assay by using it for the phenotypic characterisation of two P. falciparum conditional mutants displaying reduced fitness in vitro.


Subject(s)
Hemolytic Plaque Technique/methods , Malaria, Falciparum/parasitology , Parasites/isolation & purification , Plasmodium falciparum/isolation & purification , Animals , Erythrocytes/parasitology , Humans , Life Cycle Stages , Merozoite Surface Protein 1/metabolism , Mutation/genetics , Phenotype , Plasmodium falciparum/growth & development
8.
Sci Rep ; 6: 21800, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26892670

ABSTRACT

Conditional genome engineering in the human malaria pathogen Plasmodium falciparum remains highly challenging. Here we describe a strategy for facile and rapid functional analysis of genes using an approach based on the Cre/lox system and tailored for organisms with short and few introns. Our method allows the conditional, site-specific removal of genomic sequences of essential and non-essential genes by placing loxP sites into a short synthetic intron to produce a module (loxPint) can be placed anywhere in open reading frames without compromising protein expression. When duplicated, the loxPint module serves as an intragenic recombineering point that can be used for the fusion of gene elements to reporters or the conditional introduction of point mutations. We demonstrate the robustness and versatility of the system by targeting the P. falciparum merozoite surface protein 1 gene (msp1), which has previously proven refractory to genetic interrogation, and the parasite exported kinase FIKK10.1.


Subject(s)
Mutagenesis, Site-Directed/methods , Plasmodium falciparum/genetics , Base Sequence , Genome, Protozoan , Integrases/genetics , Introns
9.
Cell Host Microbe ; 18(4): 433-44, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468747

ABSTRACT

The malaria parasite Plasmodium falciparum replicates within erythrocytes, producing progeny merozoites that are released from infected cells via a poorly understood process called egress. The most abundant merozoite surface protein, MSP1, is synthesized as a large precursor that undergoes proteolytic maturation by the parasite protease SUB1 just prior to egress. The function of MSP1 and its processing are unknown. Here we show that SUB1-mediated processing of MSP1 is important for parasite viability. Processing modifies the secondary structure of MSP1 and activates its capacity to bind spectrin, a molecular scaffold protein that is the major component of the host erythrocyte cytoskeleton. Parasites expressing an inefficiently processed MSP1 mutant show delayed egress, and merozoites lacking surface-bound MSP1 display a severe egress defect. Our results indicate that interactions between SUB1-processed merozoite surface MSP1 and the spectrin network of the erythrocyte cytoskeleton facilitate host erythrocyte rupture to enable parasite egress.


Subject(s)
Erythrocytes/parasitology , Merozoite Surface Protein 1/metabolism , Merozoites/physiology , Plasmodium falciparum/physiology , Protein Processing, Post-Translational , Protozoan Proteins/metabolism , Spectrin/metabolism , Subtilisins/metabolism , Host-Pathogen Interactions , Humans , Merozoite Surface Protein 1/chemistry , Merozoites/enzymology , Models, Biological , Plasmodium falciparum/enzymology , Protein Binding , Protein Conformation , Proteolysis
10.
PLoS One ; 10(6): e0130356, 2015.
Article in English | MEDLINE | ID: mdl-26090798

ABSTRACT

The functional characterisation of essential genes in apicomplexan parasites, such as Toxoplasma gondii or Plasmodium falciparum, relies on conditional mutagenesis systems. Here we present a novel strategy based on U1 snRNP-mediated gene silencing. U1 snRNP is critical in pre-mRNA splicing by defining the exon-intron boundaries. When a U1 recognition site is placed into the 3'-terminal exon or adjacent to the termination codon, pre-mRNA is cleaved at the 3'-end and degraded, leading to an efficient knockdown of the gene of interest (GOI). Here we describe a simple method that combines endogenous tagging with DiCre-mediated positioning of U1 recognition sites adjacent to the termination codon of the GOI which leads to a conditional knockdown of the GOI upon rapamycin-induction. Specific knockdown mutants of the reporter gene GFP and several endogenous genes of T. gondii including the clathrin heavy chain gene 1 (chc1), the vacuolar protein sorting gene 26 (vps26), and the dynamin-related protein C gene (drpC) were silenced using this approach and demonstrate the potential of this technology. We also discuss advantages and disadvantages of this method in comparison to other technologies in more detail.


Subject(s)
Gene Silencing , Ribonucleoprotein, U1 Small Nuclear/genetics , Toxoplasma/genetics , Base Sequence , Binding Sites , Clathrin Heavy Chains/genetics , Exons , Gene Expression , Gene Targeting , Genes, Reporter , Genetic Loci , Genetic Vectors/genetics , Homologous Recombination , Molecular Sequence Data , Nucleic Acid Conformation , Nucleotide Motifs , Plasmodium falciparum/genetics , Protein Binding , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , Ribonucleoprotein, U1 Small Nuclear/chemistry , Ribonucleoprotein, U1 Small Nuclear/metabolism , Sequence Alignment
11.
Mol Microbiol ; 88(4): 687-701, 2013 May.
Article in English | MEDLINE | ID: mdl-23489321

ABSTRACT

Asexual blood stages of the malaria parasite, which cause all the pathology associated with malaria, can readily be genetically modified by homologous recombination, enabling the functional study of parasite genes that are not essential in this part of the life cycle. However, no widely applicable method for conditional mutagenesis of essential asexual blood-stage malarial genes is available, hindering their functional analysis. We report the application of the DiCre conditional recombinase system to Plasmodium falciparum, the causative agent of the most dangerous form of malaria. We show that DiCre can be used to obtain rapid, highly regulated site-specific recombination in P. falciparum, capable of excising loxP-flanked sequences from a genomic locus with close to 100% efficiency within the time-span of a single erythrocytic growth cycle. DiCre-mediated deletion of the SERA5 3' UTR failed to reduce expression of the gene due to the existence of alternative cryptic polyadenylation sites within the modified locus. However, we successfully used the system to recycle the most widely used drug resistance marker for P. falciparum, human dihydrofolate reductase, in the process producing constitutively DiCre-expressing P. falciparum clones that have broad utility for the functional analysis of essential asexual blood-stage parasite genes.


Subject(s)
Gene Deletion , Genetics, Microbial/methods , Integrases/metabolism , Molecular Biology/methods , Parasitology/methods , Plasmodium falciparum/genetics , Gene Expression , Genes, Protozoan , Integrases/genetics , Plasmodium falciparum/growth & development , Recombination, Genetic
12.
Mol Biochem Parasitol ; 179(1): 8-17, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21600245

ABSTRACT

Plasmodium falciparum enolase (Pfeno) localizes to the cytosol, nucleus, cell membrane and cytoskeletal elements, suggesting multiple non-glycolytic functions for this protein. Our recent observation of association of enolase with the food vacuole (FV) in immuno-gold electron microscopic images of P. falciparum raised the possibility for yet another moonlighting function for this protein. Here we provide additional support for this localization by demonstrating the presence of Pfeno in purified FVs by immunoblotting. To examine the potential functional role of FV-associated Pfeno, we assessed the ability of Pfeno to complement a mutant Saccharomyces cervisiae strain deficient in enolase activity. In this strain (Tetr-Eno2), the enolase 1 gene is deleted and expression of the enolase 2 gene is under the control of a tetracycline repressible promoter. Enolase deficiency in this strain was previously shown to cause growth retardation, vacuolar fragmentation and altered expression of certain vacuolar proteins. Expression of Pfeno in the enolase-deficient yeast strain restored all three phenotypic effects. However, transformation of Tetr-eno2 with an enzymatically active, monomeric mutant form of Pfeno (Δ(5)Pfeno) fully restored cell growth, but only partially rescued the fragmented vacuolar phenotype, suggesting that the dimeric structure of Pfeno is required for the optimal vacuolar functions. Bioinformatic searches revealed the presence of Plasmodium orthologs of several yeast vacuolar proteins that are predicted to form complexes with Pfeno. Together, these observations raise the possibility that association of Pfeno with food vacuole in Plasmodium may have physiological function(s).


Subject(s)
Phosphopyruvate Hydratase/metabolism , Plasmodium falciparum/enzymology , Vacuoles/chemistry , Gene Deletion , Genetic Complementation Test , Immunoblotting , Phosphopyruvate Hydratase/genetics , Plasmodium falciparum/chemistry , Saccharomyces/genetics , Saccharomyces/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...