Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Parasitol Int ; 86: 102479, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34628068

ABSTRACT

Plasmodium, the causative agents of malaria, are obligate intracellular organisms. In humans, pathogenesis is caused by the blood stage parasite, which multiplies within erythrocytes, thus erythrocyte invasion is an essential developmental step. Merozoite form parasites released into the blood stream coordinately secrets a panel of proteins from the microneme secretory organelles for gliding motility, establishment of a tight junction with a target naive erythrocyte, and subsequent internalization. A protein identified in Toxoplasma gondii facilitates microneme fusion with the plasma membrane for exocytosis; namely, acylated pleckstrin homology domain-containing protein (APH). To obtain insight into the differential microneme discharge by malaria parasites, in this study we analyzed the consequences of APH deletion in the rodent malaria model, Plasmodium yoelii, using a DiCre-based inducible knockout method. We found that APH deletion resulted in a reduction in parasite asexual growth and erythrocyte invasion, with some parasites retaining the ability to invade and grow without APH. APH deletion impaired the secretion of microneme proteins, MTRAP and AMA1, and upon contact with erythrocytes the secretion of MTRAP, but not AMA1, was observed. APH-deleted merozoites were able to attach to and deform erythrocytes, consistent with the observed MTRAP secretion. Tight junctions were formed, but echinocytosis after merozoite internalization into erythrocytes was significantly reduced, consistent with the observed absence of AMA1 secretion. Together with our observation that APH largely colocalized with MTRAP, but less with AMA1, we propose that APH is directly involved in MTRAP secretion; whereas any role of APH in AMA1 secretion is indirect in Plasmodium.


Subject(s)
Antigens, Protozoan/genetics , Gene Deletion , Plasmodium yoelii/genetics , Protozoan Proteins/genetics , Acylation , Antigens, Protozoan/metabolism , Plasmodium yoelii/metabolism , Protozoan Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article in English | MEDLINE | ID: mdl-34819379

ABSTRACT

Plasmodium malaria parasites are obligate intracellular protozoans that use a unique form of locomotion, termed gliding motility, to move through host tissues and invade cells. The process is substrate dependent and powered by an actomyosin motor that drives the posterior translocation of extracellular adhesins which, in turn, propel the parasite forward. Gliding motility is essential for tissue translocation in the sporozoite and ookinete stages; however, the short-lived erythrocyte-invading merozoite stage has never been observed to undergo gliding movement. Here we show Plasmodium merozoites possess the ability to undergo gliding motility in vitro and that this mechanism is likely an important precursor step for successful parasite invasion. We demonstrate that two human infective species, Plasmodium falciparum and Plasmodium knowlesi, have distinct merozoite motility profiles which may reflect distinct invasion strategies. Additionally, we develop and validate a higher throughput assay to evaluate the effects of genetic and pharmacological perturbations on both the molecular motor and the complex signaling cascade that regulates motility in merozoites. The discovery of merozoite motility provides a model to study the glideosome and adds a dimension for work aiming to develop treatments targeting the blood stage invasion pathways.


Subject(s)
Erythrocytes/parasitology , Merozoites/physiology , Plasmodium falciparum/genetics , Plasmodium/metabolism , Protozoan Proteins/metabolism , Sporozoites/physiology , Actin Cytoskeleton/metabolism , Actomyosin/chemistry , Animals , Erythrocytes/cytology , Human Umbilical Vein Endothelial Cells , Humans , Inhibitory Concentration 50 , Locomotion , Membrane Proteins/metabolism , Signal Transduction
3.
Parasitol Int ; 85: 102435, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34390881

ABSTRACT

Malaria remains a heavy global burden on human health, and it is important to understand the molecular and cellular biology of the parasite to find targets for drug and vaccine development. The mouse malaria model is an essential tool to characterize the function of identified molecules; however, robust technologies for targeted gene deletions are still poorly developed for the widely used rodent malaria parasite, Plasmodium yoelii. To overcome this problem, we established a DiCre-loxP inducible knockout (iKO) system in P. yoelii, which showed more than 80% excision efficacy of the target locus and more than 90% reduction of locus transcripts 24 h (one cell cycle) after RAP administration. Using this developed system, cAMP-dependent protein kinase (PKAc) was inducibly disrupted and the phenotypes of the resulting PKAc-iKO parasites were analyzed. We found that PKAc-iKO parasites showed severe growth and erythrocyte invasion defects. We also found that disruption of PKAc impaired the secretion of AMA1 in P. yoelii, in contrast to a report showing no role of PKAc in AMA1 secretion in P. falciparum. This discrepancy may be related to the difference in the timing of AMA1 distribution to the merozoite surface, which occurs just after egress for P. falciparum, but after several minutes for P. yoelii. Secretions of PyEBL, Py235, and RON2 were not affected by the disruption of PKAc in P. yoelii. PyRON2 was already secreted to the merozoite surface immediately after merozoite egress, which is inconsistent with the current model that RON2 is injected into the erythrocyte cytosol. Further investigations are required to understand the role of RON2 exposed on the merozoite surface.


Subject(s)
Antigens, Protozoan/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , Membrane Proteins/biosynthesis , Plasmodium yoelii/genetics , Protozoan Proteins/genetics , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Merozoites/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Plasmodium yoelii/enzymology , Plasmodium yoelii/metabolism , Protozoan Proteins/biosynthesis , Protozoan Proteins/metabolism
4.
Molecules ; 26(6)2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33801067

ABSTRACT

Chemical conversion of the extract of natural resources is a very attractive way to expand the chemical space to discover bioactive compounds. In order to search for new medicines to treat parasitic diseases that cause high morbidity and mortality in affected countries in the world, the ethyl acetate extract from the rhizome of Alpinia galanga (L.) has been chemically converted by epoxidation using dioxirane generated in situ. The biological activity of chemically converted extract (CCE) of A. galanga (L.) significantly increased the activity against Leishmania major up to 82.6 ± 6.2 % at 25 µg/mL (whereas 2.7 ± 0.8% for the original extract). By bioassay-guided fractionation, new phenylpropanoids (1-6) and four known compounds, hydroquinone (7), 4-hydroxy(4-hydroxyphenyl)methoxy)benzaldehyde (8), isocoumarin cis 4-hydroxymelein (9), and (2S,3S,6R,7R,9S,10S)-humulene triepoxide (10) were isolated from CCE. The structures of isolated compounds were determined by spectroscopic analyses of 1D and 2D NMR, IR, and MS spectra. The most active compound was hydroquinone (7) with IC50 = 0.37 ± 1.37 µg/mL as a substantial active principle of CCE. In addition, the new phenylpropanoid 2 (IC50 = 27.8 ± 0.34 µg/mL) also showed significant activity against L. major compared to the positive control miltefosine (IC50 = 7.47 ± 0.3 µg/mL). The activities of the isolated compounds were also evaluated against Plasmodium falciparum, Trypanosoma brucei gambisense and Trypanosoma brucei rhodeisense. Interestingly, compound 2 was selectively active against trypanosomes with potent activity. To the best of our knowledge, this is the first report on the bioactive "unnatural" natural products from the crude extract of A. galanga (L.) by chemical conversion and on its activities against causal pathogens of leishmaniasis, trypanosomiasis, and malaria.


Subject(s)
Alpinia/chemistry , Antimalarials , Plant Extracts/chemistry , Plasmodium falciparum/growth & development , Propanols , Trypanosoma brucei gambiense/growth & development , Trypanosoma brucei rhodesiense/growth & development , Antimalarials/chemistry , Antimalarials/isolation & purification , Antimalarials/pharmacology , Propanols/chemistry , Propanols/isolation & purification , Propanols/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/isolation & purification , Trypanocidal Agents/pharmacology
5.
Parasitol Int ; 83: 102358, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33901679

ABSTRACT

Plasmodium falciparum malaria parasites export several hundred proteins to the cytoplasm of infected red blood cells (RBCs) to modify the cell environment suitable for their growth. A Plasmodium translocon of exported proteins (PTEX) is necessary for both soluble and integral membrane proteins to cross the parasitophorous vacuole (PV) membrane surrounding the parasite inside the RBC. However, the molecular composition of the translocation complex for integral membrane proteins is not fully characterized, especially at the parasite plasma membrane. To examine the translocation complex, here we used mini-SURFIN4.1, consisting of a short N-terminal region, a transmembrane region, and a cytoplasmic region of an exported integral membrane protein SURFIN4.1. We found that mini-SURFIN4.1 forms a translocation intermediate complex with core PTEX components, EXP2, HSP101, and PTEX150. We also found that several proteins are exposed to the PV space, including Pf113, an uncharacterized PTEX-associated protein. We determined that Pf113 localizes in dense granules at the merozoite stage and on the parasite periphery after RBC invasion. Using an inducible translocon-clogged mini-SURFIN4.1, we found that a stable translocation intermediate complex forms at the parasite plasma membrane and contains EXP2 and a processed form of Pf113. These results suggest a potential role of Pf113 for the translocation step of mini-SURFIN4.1, providing further insights into the translocation mechanisms for parasite integral membrane proteins.


Subject(s)
Erythrocytes/parasitology , Membrane Proteins/genetics , Plasmodium falciparum/physiology , Protozoan Proteins/genetics , Animals , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Protozoan Proteins/metabolism
6.
PLoS Pathog ; 16(10): e1008917, 2020 10.
Article in English | MEDLINE | ID: mdl-33017449

ABSTRACT

Babesia bovis causes a pathogenic form of babesiosis in cattle. Following invasion of red blood cells (RBCs) the parasite extensively modifies host cell structural and mechanical properties via the export of numerous proteins. Despite their crucial role in virulence and pathogenesis, such proteins have not been comprehensively characterized in B. bovis. Here we describe the surface biotinylation of infected RBCs (iRBCs), followed by proteomic analysis. We describe a multigene family (mtm) that encodes predicted multi-transmembrane integral membrane proteins which are exported and expressed on the surface of iRBCs. One mtm gene was downregulated in blasticidin-S (BS) resistant parasites, suggesting an association with BS uptake. Induced knockdown of a novel exported protein encoded by BBOV_III004280, named VESA export-associated protein (BbVEAP), resulted in a decreased growth rate, reduced RBC surface ridge numbers, mis-localized VESA1, and abrogated cytoadhesion to endothelial cells, suggesting that BbVEAP is a novel virulence factor for B. bovis.


Subject(s)
Babesia bovis/pathogenicity , Babesiosis/parasitology , Endothelial Cells/parasitology , Erythrocytes/parasitology , Animals , Babesia bovis/genetics , Cattle , Cattle Diseases/parasitology , Membrane Proteins , Parasites/pathogenicity , Proteomics/methods , Virulence Factors/genetics
7.
Parasitol Int ; 76: 102056, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31953169

ABSTRACT

Malaria parasites proliferate by repeated invasion of and multiplication within erythrocytes in the vertebrate host. Sexually committed intraerythrocytic parasites undergo sexual stage differentiation to become gametocytes. After ingestion by the mosquito, male and female gametocytes egress from erythrocytes and fertilize within the mosquito midgut. A complex signaling pathway likely responds to environmental events to trigger gametogenesis and regulate fertilization; however, such knowledge remains limited for malaria parasites. Several pseudokinases are highly transcribed at the gametocyte stage and are possible multi-functional regulators controlling critical steps of the life cycle. Here we characterized one pseudokinase, termed PypPK1, in Plasmodium yoelii that is highly expressed in schizonts and male gametocytes. Immunofluorescence assays for parasites expressing Myc-tagged PypPK1 confirmed that PypPK1 protein is expressed in schizonts and sexual stage parasites. Transgenic ΔpPK1 parasites, in which the PypPK1 gene locus was deleted by the CRISPR/Cas9 method, showed significant growth defect and reduced virulence in mice. In the blood stage, ΔpPK1 parasites were able to egress from erythrocytes similar to wild type parasites; however, erythrocyte invasion efficacy was significantly reduced. During sexual stage development, no clear changes were seen in male and female gametocytemias as well as gametocyte egress from erythrocytes; but, the number of exflagellation centers and oocysts were significantly reduced in ΔpPK1 parasites. Taken together, PypPK1 has an important role for both erythrocyte invasion and exflagellation center formation.


Subject(s)
Erythrocytes/parasitology , Plasmodium yoelii/enzymology , Protozoan Proteins/genetics , Animals , Female , Gametogenesis , Life Cycle Stages , Male , Mice , Mice, Inbred BALB C , Plasmodium yoelii/pathogenicity , Protozoan Proteins/metabolism , Schizonts/enzymology , Schizonts/pathogenicity
8.
PLoS One ; 14(12): e0226884, 2019.
Article in English | MEDLINE | ID: mdl-31860644

ABSTRACT

Plasmodium vivax is the leading cause of malaria outside Africa and represents a significant health and economic burden on affected countries. A major obstacle for P. vivax eradication is the dormant hypnozoite liver stage that causes relapse infections and the limited antimalarial drugs that clear this stage. Advances in studying the hypnozoite and other unique biological aspects of this parasite are hampered by the lack of a continuous in vitro laboratory culture system and poor availability of molecular tools for genetic manipulation. In this study, we aim to develop molecular tools that can be used for genetic manipulation of P. vivax. A putative P. vivax centromere sequence (PvCEN) was cloned and episomal centromere based plasmids expressing a GFP marker were constructed. Centromere activity was evaluated using a rodent malaria parasite Plasmodium yoelii. A plasmid carrying PvCEN was stably maintained in asexual-stage parasites in the absence of drug pressure, and approximately 45% of the parasites retained the plasmid four weeks later. The same retention rate was observed in parasites possessing a native P. yoelii centromere (PyCEN)-based control plasmid. The segregation efficiency of the plasmid per nuclear division was > 99% in PvCEN parasites, compared to ~90% in a control parasite harboring a plasmid without a centromere. In addition, we observed a clear GFP signal in both oocysts and salivary gland sporozoites isolated from mosquitoes. In blood-stage parasites after liver stage development, GFP positivity in PvCEN parasites was comparable to control PyCEN parasites. Thus, PvCEN plasmids were maintained throughout the parasite life cycle. We also validated several P. vivax promoter activities and showed that hsp70 promoter (~1 kb) was active throughout the parasite life cycle. This is the first data for the functional characterization of a P. vivax centromere that can be used in future P. vivax biological research.


Subject(s)
Centromere/genetics , Plasmodium vivax/genetics , Plasmodium yoelii/genetics , Promoter Regions, Genetic/genetics , Animals , Chromosome Segregation/genetics , Culicidae/parasitology , Female , Green Fluorescent Proteins/metabolism , Humans , Malaria, Vivax/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Microorganisms, Genetically-Modified/genetics , Plasmids/genetics , Plasmodium yoelii/growth & development , Salivary Glands/parasitology , Sporozoites/metabolism , Tetrahydrofolate Dehydrogenase/genetics
9.
mBio ; 10(5)2019 09 17.
Article in English | MEDLINE | ID: mdl-31530668

ABSTRACT

Plasmodium falciparum has a complex life cycle that involves interaction with multiple tissues inside the human and mosquito hosts. Identification of essential genes at all different stages of the P. falciparum life cycle is urgently required for clinical development of tools for malaria control and eradication. However, the study of P. falciparum is limited by the inability to genetically modify the parasite throughout its life cycle with the currently available genetic tools. Here, we describe the detailed characterization of a new marker-free P. falciparum parasite line that expresses rapamycin-inducible Cre recombinase across the full life cycle. Using this parasite line, we were able to conditionally delete the essential invasion ligand AMA1 in three different developmental stages for the first time. We further confirm efficient gene deletion by targeting the nonessential kinase FIKK7.1.IMPORTANCE One of the major limitations in studying P. falciparum is that so far only asexual stages are amenable to rapid conditional genetic modification. The most promising drug targets and vaccine candidates, however, have been refractory to genetic modification because they are essential during the blood stage or for transmission in the mosquito vector. This leaves a major gap in our understanding of parasite proteins in most life cycle stages and hinders genetic validation of drug and vaccine targets. Here, we describe a method that supports conditional gene deletion across the P. falciparum life cycle for the first time. We demonstrate its potential by deleting essential and nonessential genes at different parasite stages, which opens up completely new avenues for the study of malaria and drug development. It may also allow the realization of novel vaccination strategies using attenuated parasites.


Subject(s)
Gene Deletion , Genes, Protozoan , Life Cycle Stages/genetics , Molecular Biology/methods , Plasmodium falciparum/genetics , Gene Knockout Techniques , Integrases/genetics , Mosquito Vectors , Phenotype , Plasmodium falciparum/enzymology , Sirolimus
10.
Parasitol Int ; 71: 186-193, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31028841

ABSTRACT

Plasmodium falciparum, an obligate intracellular protozoan parasite which causes the severe form of human malaria, exports numerous proteins to the infected red blood cell that are important for its survival and of severe pathological effect to its host. These proteins and their export mechanisms are candidates for drug and vaccine development, and among them is the Plasmodium SURFIN family of proteins. Previously we showed that the N-terminal region along with the sequence surrounding the transmembrane domain of SURFIN4.1 is essential for its export to Maurer's clefts in the red blood cell cytoplasm. We proposed that this region is recognized by a machinery responsible for protein translocation across the parasitophorous vacuole membrane surrounding the parasite. To understand the export mechanism further, we utilized a fluorescent protein-tagged mini-SURFIN4.1 consisting of the minimum essential components for export. Alanine scanning of all charged amino acids within the N-terminal region revealed that replacement of 3 glutamic acid and 2 lysine residues significantly impairs the export efficiency of this protein across the parasitophorous vacuole membrane. In addition, N-terminally Myc-tagged mini-SURFIN4.1 and mini-SURFIN4.2 with similar architectures were detected with anti-Myc antibody at Maurer's clefts, indicating that elements required for export to Maurer's clefts are conserved between SURFIN4.1 and SURFIN4.2, and that N-terminal sequences of these SURFIN members are not cleaved during export. Our results implicate a conserved nature of SURFIN export to the red blood cell, particularly an important role of multiple glutamic acid and lysine residues in the SURFIN N-terminal region.


Subject(s)
Amino Acids/chemistry , Erythrocytes/parasitology , Host-Parasite Interactions , Membrane Proteins/chemistry , Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Glutamic Acid/chemistry , Humans , Lysine/chemistry , Protein Transport
11.
J Phys Chem B ; 122(33): 7970-7977, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30067362

ABSTRACT

Plasmodium falciparum, the causative agent of malignant malaria, is insensitive to thapsigargin (TG), a well-known inhibitor of the human sarco/endoplasmic reticulum Ca2+-ATPase (SERCA). To understand the key factor causing the difference of the sensitivity, the molecular interaction of TG and each SERCA was analyzed by the fragment molecular orbital (FMO) method. While the major component of the interaction energy was the nonpolar interaction, the major difference in the molecular interaction arose from the polar interaction, namely, the hydrogen bonding interaction with a hydroxyl group of TG. Additionally, we successfully confirmed these FMO calculation results by measuring the inhibitory activity of a synthesized TG derivative. Our calculations and experiments indicated that, by replacing the hydroxyl group of TG with another functional group, the sensitivities of TG to human and P. falciparum SERCAs can be reversed. This study provides important information to develop antimalarial compounds targeting P. falciparum SERCA.


Subject(s)
Antimalarials/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/metabolism , Amino Acid Sequence , Antimalarials/chemistry , Humans , Hydrogen Bonding , Models, Molecular , Plasmodium falciparum/enzymology , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Quantum Theory , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sequence Alignment , Thapsigargin/analogs & derivatives , Thapsigargin/chemical synthesis
12.
Parasitol Int ; 67(6): 706-714, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30025976

ABSTRACT

Plasmodium malaria parasites multiply within erythrocytes and possess a repertoire of proteins whose function is to recognize and invade these vertebrate host cells. One such protein involved in erythrocyte invasion is the micronemal protein, Erythrocyte Binding-Like (EBL), which has been studied as a potential target of vaccine development in Plasmodium vivax (PvDBP) and Plasmodium falciparum (EBA-175). In the rodent malaria parasite model Plasmodium yoelii, specific substitutions in the EBL regions responsible for intracellular trafficking (17XL parasite line) or receptor recognition (17X1.1pp. parasite line), paradoxically increase invasion ability and virulence rather than abolish EBL function. Attempts to disrupt the ebl gene locus in the 17XL and 17XNL lines were unsuccessful, suggesting EBL essentiality. To understand the mechanisms behind these potentially conflicting outcomes, we generated 17XL-based transfectants in which ebl expression is suppressed with anhydrotetracycline (ATc) and investigated merozoite behavior during erythrocyte invasion. In the absence of ATc, EBL was secreted to the merozoite surface, whereas following ATc administration parasitemia was negligible in vivo. Merozoites lacking EBL were unable to invade erythrocytes in vitro, indicating that EBL has a critical role for erythrocyte invasion. Quantitative time-lapse imaging revealed that with ATc administration a significant number of merozoites were detached from the erythrocyte after the erythrocyte deformation event and no echinocytosis was observed, indicating that EBL is required for merozoites to establish an irreversible connection with erythrocytes during invasion.


Subject(s)
Antigens, Protozoan/metabolism , Erythrocytes/parasitology , Malaria/parasitology , Parasitemia/parasitology , Plasmodium yoelii/physiology , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Female , Gene Knockdown Techniques , Mice , Mice, Inbred ICR
13.
Parasitol Int ; 67(4): 481-492, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29673877

ABSTRACT

During development within the host erythrocyte malaria parasites generate nascent membranous structures which serve as a pathway for parasite protein transport to modify the host cell. The molecular basis of such membranous structures is not well understood, particularly for malaria parasites other than Plasmodium falciparum. To characterize the structural basis of protein trafficking in the Plasmodium knowlesi-infected erythrocyte, we identified a P. knowlesi ortholog of MAHRP2, a marker of the tether structure that connects membranous structures in the P. falciparum-infected erythrocyte. We show that PkMAHRP2 localizes on amorphous structures that connect Sinton Mulligan's clefts (SMC) to each other and to the erythrocyte membrane. Three dimensional reconstruction of the P. knowlesi-infected erythrocyte revealed that the SMC is a plate-like structure with swollen ends, reminiscent of the morphology of the Golgi apparatus. The PkMAHRP2-localized amorphous structures are possibly functionally equivalent to P. falciparum tether structure. These findings suggest a conservation in the ultrastructure of protein trafficking between P. falciparum and P. knowlesi.


Subject(s)
Erythrocytes/parasitology , Plasmodium knowlesi/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Erythrocytes/chemistry , Host-Parasite Interactions , Membrane Proteins/analysis , Plasmodium falciparum/chemistry , Plasmodium falciparum/metabolism , Plasmodium knowlesi/chemistry , Plasmodium knowlesi/genetics , Protein Transport , Protozoan Proteins/metabolism
14.
Parasitol Int ; 67(3): 284-293, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29353010

ABSTRACT

Individuals living in malaria endemic areas become clinically immune after multiple re-infections over time and remain infected without apparent symptoms. However, it is unclear why a long period is required to gain clinical immunity to malaria, and how such immunity is maintained. Although malaria infection is reported to induce inhibition of immune responses, studies on asymptomatic individuals living in endemic regions of malaria are relatively scarce. We conducted a cross-sectional study of immune responses in asymptomatic school children aged 4-16years living in an area where Plasmodium falciparum and Schistosoma mansoni infections are co-endemic in Kenya. Peripheral blood mononuclear cells were subjected to flow cytometric analysis and cultured to determine proliferative responses and cytokine production. The proportions of cellular subsets in children positive for P. falciparum infection at the level of microscopy were comparable to the negative children, except for a reduction in central memory-phenotype CD8+ T cells and natural killer cells. In functional studies, the production of cytokines by peripheral blood mononuclear cells in response to P. falciparum crude antigens exhibited strong heterogeneity among children. In addition, production of IL-2 in response to anti-CD3 and anti-CD28 monoclonal antibodies was significantly reduced in P. falciparum-positive children as compared to -negative children, suggesting a state of unresponsiveness. These data suggest that the quality of T cell immune responses is heterogeneous among asymptomatic children living in the endemic region of P. falciparum, and that the responses are generally suppressed by active infection with Plasmodium parasites.


Subject(s)
Asymptomatic Infections/epidemiology , Endemic Diseases , Malaria, Falciparum/epidemiology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adolescent , Animals , Antigens, Helminth/immunology , Biomphalaria , CD8-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Cross-Sectional Studies , Cytokines/biosynthesis , Female , Flow Cytometry , Humans , Immunity, Innate , Kenya/epidemiology , Killer Cells, Natural , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Malaria, Falciparum/complications , Male , Mice , Mice, Inbred ICR , Schistosoma mansoni/immunology , Schistosomiasis mansoni/complications
15.
PLoS Pathog ; 13(7): e1006447, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28704525

ABSTRACT

Identifying the genetic determinants of phenotypes that impact disease severity is of fundamental importance for the design of new interventions against malaria. Here we present a rapid genome-wide approach capable of identifying multiple genetic drivers of medically relevant phenotypes within malaria parasites via a single experiment at single gene or allele resolution. In a proof of principle study, we found that a previously undescribed single nucleotide polymorphism in the binding domain of the erythrocyte binding like protein (EBL) conferred a dramatic change in red blood cell invasion in mutant rodent malaria parasites Plasmodium yoelii. In the same experiment, we implicated merozoite surface protein 1 (MSP1) and other polymorphic proteins, as the major targets of strain-specific immunity. Using allelic replacement, we provide functional validation of the substitution in the EBL gene controlling the growth rate in the blood stages of the parasites.


Subject(s)
Antigens, Protozoan/genetics , Malaria/immunology , Malaria/parasitology , Merozoite Surface Protein 1/genetics , Plasmodium yoelii/genetics , Plasmodium yoelii/pathogenicity , Protozoan Proteins/genetics , Receptors, Cell Surface/genetics , Antigens, Protozoan/metabolism , Erythrocytes/parasitology , Host-Parasite Interactions , Humans , Immunity , Malaria/genetics , Merozoite Surface Protein 1/metabolism , Plasmodium yoelii/growth & development , Plasmodium yoelii/metabolism , Polymorphism, Single Nucleotide , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Virulence
16.
Malar J ; 16(1): 98, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28253868

ABSTRACT

BACKGROUND: Plasmodium falciparum SURFIN4.1 is a putative ligand expressed on the merozoite and likely on the infected red blood cell, whose gene was suggested to be under directional selection in the eastern Kenyan population, but under balancing selection in the Thai population. To understand this difference, surf 4.1 sequences of western Kenyan P. falciparum isolates were analysed. Frameshift mutations and copy number variation (CNV) were also examined for the parasites from western Kenya and Thailand. RESULTS: Positively significant departures from neutral expectations were detected on the surf 4.1 region encoding C-terminus of the variable region 2 (Var2) by 3 population-based tests in the western Kenyan population as similar in the Thai population, which was not covered by the previous analysis for eastern Kenyan population. Significant excess of non-synonymous substitutions per nonsynonymous site over synonymous substitutions per synonymous site was also detected in the Var2 region. Negatively significant departures from neutral expectations was detected on the region encoding Var1 C-terminus consistent to the previous observation in the eastern Kenyan population. Parasites possessing a frameshift mutation resulting a product without intracellular Trp-rich (WR) domains were 22/23 in western Kenya and 22/36 in Thailand. More than one copy of surf 4.1 gene was detected in western Kenya (4/24), but no CNV was found in Thailand (0/36). CONCLUSIONS: The authors infer that the high polymorphism of SURFIN4.1 Var2 C-terminus in both Kenyan and Thai populations were shaped-up by diversifying selection and maintained by balancing selection. These phenomena were most likely driven by immunological pressure. Whereas the SURFIN4.1 Var1 C-terminus is suggested to be under directional selection consistent to the previous report for the eastern Kenyan population. Most western Kenyan isolates possess a frameshift mutation that would limit the expression of SURFIN4.1 on the merozoite, but only 60% of Thai isolates possess this frameshift, which would affect the level and type of the selection pressure against this protein as seen in the two extremities of Tajima's D values for Var1 C-terminus between Kenyan and Thai populations. CNV observed in Kenyan isolates may be a consequence of this frameshift mutation to increase benefits on the merozoite surface.


Subject(s)
Frameshift Mutation , Gene Dosage , Membrane Proteins/genetics , Plasmodium falciparum/genetics , Polymorphism, Genetic , Protozoan Proteins/genetics , Selection, Genetic , Kenya , Plasmodium falciparum/isolation & purification , Sequence Analysis, DNA , Thailand
17.
PLoS One ; 11(10): e0164272, 2016.
Article in English | MEDLINE | ID: mdl-27732628

ABSTRACT

The malaria parasite, Plasmodium, exports protein products to the infected erythrocyte to introduce modifications necessary for the establishment of nutrient acquisition and surface display of host interaction ligands. Erythrocyte remodeling impacts parasite virulence and disease pathology and is well documented for the human malaria parasite Plasmodium falciparum, but has been less described for other Plasmodium species. For P. falciparum, the exported protein skeleton-binding protein 1 (PfSBP1) is involved in the trafficking of erythrocyte surface ligands and localized to membranous structures within the infected erythrocyte, termed Maurer's clefts. In this study, we analyzed SBP1 orthologs across the Plasmodium genus by BLAST analysis and conserved gene synteny, which were also recently described by de Niz et al. (2016). To evaluate the localization of an SBP1 ortholog, we utilized the zoonotic malaria parasite, Plasmodium knowlesi. Immunofluorescence assay of transgenic P. knowlesi parasites expressing epitope-tagged recombinant PkSBP1 revealed a punctate staining pattern reminiscent of Maurer's clefts, following infection of either monkey or human erythrocytes. The recombinant PkSBP1-positive puncta co-localized with Giemsa-stained structures, known as 'Sinton and Mulligan' stipplings. Immunoelectron microscopy also showed that recombinant PkSBP1 localizes within or on the membranous structures akin to the Maurer's clefts. The recombinant PkSBP1 expressed in P. falciparum-infected erythrocytes co-localized with PfSBP1 at the Maurer's clefts, indicating an analogous trafficking pattern. A member of the P. knowlesi 2TM protein family was also expressed and localized to membranous structures in infected monkey erythrocytes. These results suggest that the trafficking machinery and induced erythrocyte cellular structures of P. knowlesi are similar following infection of both monkey and human erythrocytes, and are conserved with P. falciparum.


Subject(s)
Carrier Proteins/metabolism , Erythrocytes/pathology , Erythrocytes/parasitology , Malaria/pathology , Malaria/parasitology , Plasmodium knowlesi/physiology , Protozoan Proteins/metabolism , Animals , Carrier Proteins/analysis , Cytoplasm/metabolism , Cytoplasm/parasitology , Cytoplasm/pathology , Erythrocytes/metabolism , Haplorhini , Humans , Malaria/metabolism , Protein Transport , Protozoan Proteins/analysis
18.
Malar J ; 15: 323, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27316546

ABSTRACT

BACKGROUND: Rab5 GTPase regulates membrane trafficking between the plasma membrane and endosomes and harbours a conserved C-terminal isoprenyl modification that is necessary for membrane recruitment. Plasmodium falciparum encodes three Rab5 isotypes, and one of these, Rab5b (PfRab5b), lacks the C-terminal modification but possesses the N-terminal myristoylation motif. PfRab5b was reported to localize to the parasite periphery. However, the trafficking pathway regulated by PfRab5b is unknown. METHODS: A complementation analysis of Rab5 isotypes was performed in Plasmodium berghei. A constitutively active PfRab5b mutant was expressed under the regulation of a ligand-dependent destabilization domain (DD)-tag system in P. falciparum. The localization of PfRab5b was evaluated after removing the ligand followed by selective permeabilization of the membrane with different detergents. Furthermore, P. falciparum N-terminally myristoylated adenylate kinase 2 (PfAK2) was co-expressed with PfRab5b, and trafficking of PfAK2 to the parasitophorous vacuole membrane was examined by confocal microscopy. RESULTS: PfRab5b complemented the function of PbRab5b, however, the conventional C-terminally isoprenylated Rab5, PbRab5a or PbRab5c, did not. The constitutively active PfRab5b mutant localized to the cytosol of the parasite and the tubovesicular network (TVN), a region that extends from the parasitophorous vacuole membrane (PVM) in infected red blood cells (iRBCs). By removing the DD-ligand, parasite cytosolic PfRab5b signal disappeared and a punctate structure adjacent to the endoplasmic reticulum (ER) and parasite periphery accumulated. The peripheral PfRab5b was sensitive to extracellular proteolysis after treatment with streptolysin O, which selectively permeabilizes the red blood cell plasma membrane, indicating that PfRab5b localized on the iRBC cytoplasmic face of the TVN. Transport of PfAK2 to the PVM was abrogated by overexpression of PfRab5b, and PfAK2 accumulated in the punctate structure together with PfRab5b. CONCLUSION: N-myristoylated Plasmodium Rab5b plays a role that is distinct from that of conventional mammalian Rab5 isotypes. PfRab5b localizes to a compartment close to the ER, translocated to the lumen of the organelle, and co-localizes with PfAK2. PfRab5b and PfAK2 are then transported to the TVN, and PfRab5b localizes on the iRBC cytoplasmic face of TVN. These data demonstrate that PfRab5b is transported from the parasite cytosol to TVN together with N-myristoylated PfAK2 via an uncharacterized membrane-trafficking pathway.


Subject(s)
Adenylate Kinase/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Plasmodium berghei/enzymology , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Adenylate Kinase/genetics , Humans , Plasmodium berghei/genetics , Plasmodium falciparum/genetics , rab5 GTP-Binding Proteins/genetics
19.
Sci Rep ; 6: 23454, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27006284

ABSTRACT

Calcium (Ca(2+))-mediated signaling is a conserved mechanism in eukaryotes, including the human malaria parasite, Plasmodium falciparum. Due to its small size (<10 µm) measurement of intracellular Ca(2+) in Plasmodium is technically challenging, and thus Ca(2+) regulation in this human pathogen is not well understood. Here we analyze Ca(2+) homeostasis via a new approach using transgenic P. falciparum expressing the Ca(2+) sensor yellow cameleon (YC)-Nano. We found that cytosolic Ca(2+) concentration is maintained at low levels only during the intraerythrocytic trophozoite stage (30 nM), and is increased in the other blood stages (>300 nM). We determined that the mammalian SERCA inhibitor thapsigargin and antimalarial dihydroartemisinin did not perturb SERCA activity. The change of the cytosolic Ca(2+) level in P. falciparum was additionally detectable by flow cytometry. Thus, we propose that the developed YC-Nano-based system is useful to study Ca(2+) signaling in P. falciparum and is applicable for drug screening.


Subject(s)
Biosensing Techniques/methods , Calcium-Binding Proteins/metabolism , Calcium/analysis , Erythrocytes/parasitology , Plasmodium falciparum/genetics , Artemisinins/pharmacology , Calcium/metabolism , Calcium Signaling , Calcium-Binding Proteins/genetics , Cells, Cultured , Cytosol/chemistry , Homeostasis , Humans , Organisms, Genetically Modified , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/pharmacology , Trophozoites/chemistry
20.
Mol Biochem Parasitol ; 204(1): 26-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26684675

ABSTRACT

Malaria symptoms and pathogenesis are caused by blood stage parasite burdens of Plasmodium spp., for which invasion of red blood cells (RBCs) by merozoites is essential. Successful targeting by either drugs or vaccines directed against the whole merozoite or its antigens during its transient extracellular status would contribute to malaria control by impeding RBC invasion. To understand merozoite invasion biology and mechanisms, it is desired to obtain merozoites that retain their invasion activity in vitro. Accordingly, methods have been developed to isolate invasive Plasmodium knowlesi and Plasmodium falciparum merozoites. Rodent malaria parasite models offer ease in laboratory maintenance and experimental genetic modifications; however, no methods have been reported regarding isolation of high numbers of invasive rodent malaria merozoites. In this study, Plasmodium yoelii-infected RBCs were obtained from infected mice, and mature schizont-infected RBCs enriched via Histodenz™ density gradients. Merozoites retaining invasion activity were then isolated by passing the preparations through a filter membrane. RBC-invaded parasites developed to mature stages in vitro in a synchronous manner. Isolated merozoites were evaluated for retention of invasion activity following storage at different temperatures prior to incubation with uninfected mouse RBCs. Isolated merozoites retained their invasion activity 4h after isolation at 10 or 15 °C, whereas their invasion activity reduced to 0-10% within 30 min when incubated on ice or at 37 °C prior to RBC invasion assay. Images of merozoites at successive steps during RBC invasion were captured by light and transmission electron microscopy. Synthetic peptides derived from the amino acid sequence of the P. yoelii invasion protein RON2 efficiently inhibited RBC invasion. The developed method to isolate and keep invasive P. yoelii merozoites for up to 4h is a powerful tool to study the RBC invasion biology of this parasite. This method provides an important platform to evaluate the mode of action of drugs and vaccine candidates targeting the RBC invasion steps using rodent malaria model.


Subject(s)
Antimalarials/pharmacology , Malaria/parasitology , Plasmodium yoelii/drug effects , Plasmodium yoelii/isolation & purification , Amino Acid Sequence , Animals , Disease Models, Animal , Erythrocytes/parasitology , Female , Merozoites/drug effects , Merozoites/growth & development , Mice , Mice, Inbred ICR , Microscopy, Electron, Transmission , Molecular Sequence Data , Peptides/chemistry , Peptides/pharmacology , Plasmodium yoelii/pathogenicity , Protozoan Proteins/chemistry , Protozoan Proteins/pharmacology , Spores
SELECTION OF CITATIONS
SEARCH DETAIL
...