Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.482
Filtrar
1.
mBio ; 15(5): e0285023, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38564676

RESUMO

Condensin I is a pentameric complex that regulates the mitotic chromosome assembly in eukaryotes. The kleisin subunit CAP-H of the condensin I complex acts as a linchpin to maintain the structural integrity and loading of this complex on mitotic chromosomes. This complex is present in all eukaryotes and has recently been identified in Plasmodium spp. However, how this complex is assembled and whether the kleisin subunit is critical for this complex in these parasites are yet to be explored. To examine the role of PfCAP-H during cell division within erythrocytes, we generated an inducible PfCAP-H knockout parasite. We find that PfCAP-H is dynamically expressed during mitosis with the peak expression at the metaphase plate. PfCAP-H interacts with PfCAP-G and is a non-SMC member of the condensin I complex. Notably, the absence of PfCAP-H does not alter the expression of PfCAP-G but affects its localization at the mitotic chromosomes. While mitotic spindle assembly is intact in PfCAP-H-deficient parasites, duplicated centrosomes remain clustered over the mass of unsegmented nuclei with failed karyokinesis. This failure leads to the formation of an abnormal nuclear mass, while cytokinesis occurs normally. Altogether, our data suggest that PfCAP-H plays a crucial role in maintaining the structural integrity of the condensin I complex on the mitotic chromosomes and is essential for the asexual development of malarial parasites. IMPORTANCE: Mitosis is a fundamental process for Plasmodium parasites, which plays a vital role in their survival within two distinct hosts-human and Anopheles mosquitoes. Despite its great significance, our comprehension of mitosis and its regulation remains limited. In eukaryotes, mitosis is regulated by one of the pivotal complexes known as condensin complexes. The condensin complexes are responsible for chromosome condensation, ensuring the faithful distribution of genetic material to daughter cells. While condensin complexes have recently been identified in Plasmodium spp., our understanding of how this complex is assembled and its precise functions during the blood stage development of Plasmodium falciparum remains largely unexplored. In this study, we investigate the role of a central protein, PfCAP-H, during the blood stage development of P. falciparum. Our findings reveal that PfCAP-H is essential and plays a pivotal role in upholding the structure of condensin I and facilitating karyokinesis.


Assuntos
Adenosina Trifosfatases , Proteínas de Ligação a DNA , Mitose , Complexos Multiproteicos , Plasmodium falciparum , Proteínas de Protozoários , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/genética , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Plasmodium falciparum/crescimento & desenvolvimento , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Eritrócitos/parasitologia , Técnicas de Inativação de Genes , Humanos
2.
Cell Rep ; 43(4): 114012, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38573856

RESUMO

Plasmodium falciparum is a human-adapted apicomplexan parasite that causes the most dangerous form of malaria. P. falciparum cysteine-rich protective antigen (PfCyRPA) is an invasion complex protein essential for erythrocyte invasion. The precise role of PfCyRPA in this process has not been resolved. Here, we show that PfCyRPA is a lectin targeting glycans terminating with α2-6-linked N-acetylneuraminic acid (Neu5Ac). PfCyRPA has a >50-fold binding preference for human, α2-6-linked Neu5Ac over non-human, α2-6-linked N-glycolylneuraminic acid. PfCyRPA lectin sites were predicted by molecular modeling and validated by mutagenesis studies. Transgenic parasite lines expressing endogenous PfCyRPA with single amino acid exchange mutants indicated that the lectin activity of PfCyRPA has an important role in parasite invasion. Blocking PfCyRPA lectin activity with small molecules or with lectin-site-specific monoclonal antibodies can inhibit blood-stage parasite multiplication. Therefore, targeting PfCyRPA lectin activity with drugs, immunotherapy, or a vaccine-primed immune response is a promising strategy to prevent and treat malaria.


Assuntos
Eritrócitos , Plasmodium falciparum , Polissacarídeos , Proteínas de Protozoários , Humanos , Antígenos de Protozoários/metabolismo , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Eritrócitos/parasitologia , Eritrócitos/metabolismo , Lectinas/metabolismo , Lectinas/genética , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética
3.
Front Cell Infect Microbiol ; 14: 1304839, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38572319

RESUMO

Background: Chemotherapies for malaria and babesiosis frequently succumb to the emergence of pathogen-related drug-resistance. Host-targeted therapies are thought to be less susceptible to resistance but are seldom considered for treatment of these diseases. Methods: Our overall objective was to systematically assess small molecules for host cell-targeting activity to restrict proliferation of intracellular parasites. We carried out a literature survey to identify small molecules annotated for host factors implicated in Plasmodium falciparum infection. Alongside P. falciparum, we implemented in vitro parasite susceptibility assays also in the zoonotic parasite Plasmodium knowlesi and the veterinary parasite Babesia divergens. We additionally carried out assays to test directly for action on RBCs apart from the parasites. To distinguish specific host-targeting antiparasitic activity from erythrotoxicity, we measured phosphatidylserine exposure and hemolysis stimulated by small molecules in uninfected RBCs. Results: We identified diverse RBC target-annotated inhibitors with Plasmodium-specific, Babesia-specific, and broad-spectrum antiparasitic activity. The anticancer MEK-targeting drug trametinib is shown here to act with submicromolar activity to block proliferation of Plasmodium spp. in RBCs. Some inhibitors exhibit antimalarial activity with transient exposure to RBCs prior to infection with parasites, providing evidence for host-targeting activity distinct from direct inhibition of the parasite. Conclusions: We report here characterization of small molecules for antiproliferative and host cell-targeting activity for malaria and babesiosis parasites. This resource is relevant for assessment of physiological RBC-parasite interactions and may inform drug development and repurposing efforts.


Assuntos
Antimaláricos , Babesia , Babesiose , Malária Falciparum , Malária , Parasitos , Plasmodium , Animais , Humanos , Babesiose/tratamento farmacológico , Malária/parasitologia , Eritrócitos/parasitologia , Antimaláricos/farmacologia , Plasmodium falciparum
4.
Vet Parasitol ; 326: 110081, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38113611

RESUMO

B. bovis invasion of bovine erythrocytes requires tight junction formation involving AMA-1/RON2 complex interaction. RON2 has been considered a vaccine candidate since antibodies targeting the protein can inhibit parasite invasion of target cells; however, the mechanism controlling B. bovis RON2 interaction with red blood cells is not yet fully understood. This study was thus aimed at identifying B. bovis RON2 protein regions associated with interaction with bovine erythrocytes. Natural selection analysis of the ron2 gene identified predominantly negative selection signals in the C-terminal region. Interestingly, protein-cell and competition assays highlighted the RON2-C region's role in peptide 42918-mediated erythrocyte binding, probably to a sialoglycoprotein receptor. This peptide (1218SFIMVKPPALHCVLKPVETL1237) lies within an intrinsically disordered region of the RON2 secondary structure flanked by two helical residues. The study provides, for the first time, valuable insights into RON2's role in interaction with its target cells. Future studies are required for studying the peptide's potential as an anti-B. bovis vaccine component.


Assuntos
Babesia bovis , Vacinas , Animais , Bovinos , Epitopos , Proteínas de Protozoários/metabolismo , Peptídeos , Eritrócitos/parasitologia
5.
BMC Biol ; 21(1): 279, 2023 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-38049852

RESUMO

BACKGROUND: The rapid reproduction of malaria parasites requires proper iron uptake. However, the process of iron absorption by parasites is rarely studied. Divalent metal transporter (DMT1) is a critical iron transporter responsible for uptaking iron. A homolog of human DMT1 exists in the malaria parasite genome, which in Plasmodium yoelii is hereafter named PyDMT1. RESULTS: PyDMT1 knockout appears to be lethal. Surprisingly, despite dwelling in an iron-rich environment, the parasite cannot afford to lose even partial expression of PyDMT1; PyDMT1 hypomorphs were associated with severe growth defects and quick loss of pathogenicity. Iron supplementation could completely suppress the defect of the PyDMT1 hypomorph during in vitro culturing. Genetic manipulation through host ferritin (Fth1) knockout to increase intracellular iron levels enforced significant growth inhibition in vivo on the normal parasites but not the mutant. In vitro culturing with isolated ferritin knockout mouse erythrocytes completely rescued PyDMT1-hypomorph parasites. CONCLUSION: A critical iron requirement of malaria parasites at the blood stage as mediated by this newly identified iron importer PyDMT1, and the iron homeostasis in malarial parasites is finely tuned. Tipping the iron balance between the parasite and host will efficiently kill the pathogenicity of the parasite. Lastly, PyDMT1 hypomorph parasites were less sensitive to the action of artemisinin.


Assuntos
Malária , Plasmodium yoelii , Animais , Camundongos , Humanos , Ferro/metabolismo , Ferritinas/genética , Ferritinas/metabolismo , Transporte Biológico , Proteínas de Membrana Transportadoras/metabolismo , Eritrócitos/parasitologia
6.
Cell Rep ; 42(10): 113253, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37819760

RESUMO

Sequestration of Plasmodium falciparum-infected erythrocytes (IEs) in the brain microcirculation is a hallmark of cerebral malaria (CM), which leads to endothelial activation, brain swelling, and death. Here, we probed CM inflammation in a perfusable 3D human brain microvessel model. 3D brain microvessels supported in vivo-like capacities for parasite binding and maturation in situ, leading to a distinct inflammatory response from the pro-inflammatory cytokine tumor necrosis factor α (TNF-α). By combining transcriptional analysis, imaging, and leukocyte perfusion, we showed that whereas TNF-α promotes a reversible inflammatory phenotype with widespread leukocyte recruitment, parasites induce unique stress response pathways and cause localized cell adhesivity changes, focal endothelial disruptions, and apoptosis. Furthermore, parasites modified the temporal kinetics of the TNF transcriptional response, suggesting augmented inflammatory damage with the two sequential stimuli. Our findings offer mechanistic insights into CM biology in a 3D brain microvessel mimetic platform and suggest that multiple events intersect to promote brain barrier inflammation in CM.


Assuntos
Malária Cerebral , Malária Falciparum , Humanos , Fator de Necrose Tumoral alfa , Encéfalo/patologia , Plasmodium falciparum/genética , Inflamação/patologia , Microvasos/patologia , Eritrócitos/parasitologia , Malária Falciparum/parasitologia
7.
Blood ; 142(23): 2016-2028, 2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-37832027

RESUMO

The malaria parasite Plasmodium falciparum invades and replicates asexually within human erythrocytes. CD44 expressed on erythrocytes was previously identified as an important host factor for P falciparum infection through a forward genetic screen, but little is known about its regulation or function in these cells, nor how it may be used by the parasite. We found that CD44 can be efficiently deleted from primary human hematopoietic stem cells using CRISPR/Cas9 genome editing, and that the efficiency of ex vivo erythropoiesis to enucleated cultured red blood cells (cRBCs) is not affected by lack of CD44. However, the rate of P falciparum invasion was reduced in CD44-null cRBCs relative to isogenic wild-type control cells, validating CD44 as an important host factor for this parasite. We identified 2 P falciparum invasion ligands as binding partners for CD44, erythrocyte binding antigen 175 (EBA-175) and EBA-140 and demonstrated that their ability to bind to human erythrocytes relies primarily on their canonical receptors, glycophorin A and glycophorin C, respectively. We further show that EBA-175 induces phosphorylation of erythrocyte cytoskeletal proteins in a CD44-dependent manner. Our findings support a model in which P falciparum exploits CD44 as a coreceptor during invasion of human erythrocytes, stimulating CD44-dependent phosphorylation of host cytoskeletal proteins that alter host cell deformability and facilitate parasite entry.


Assuntos
Eritrócitos , Malária Falciparum , Plasmodium falciparum , Humanos , Antígenos de Protozoários/genética , Antígenos de Protozoários/metabolismo , Proteínas do Citoesqueleto , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Receptores de Hialuronatos/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Ligação Proteica , Proteínas de Protozoários/metabolismo
8.
Elife ; 122023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37737226

RESUMO

Reverse genetics is key to understanding protein function, but the mechanistic connection between a gene of interest and the observed phenotype is not always clear. Here we describe the use of proximity labeling using TurboID and site-specific quantification of biotinylated peptides to measure changes to the local protein environment of selected targets upon perturbation. We apply this technique, which we call PerTurboID, to understand how the Plasmodium falciparum-exported kinase, FIKK4.1, regulates the function of the major virulence factor of the malaria-causing parasite, PfEMP1. We generated independent TurboID fusions of two proteins that are predicted substrates of FIKK4.1 in a FIKK4.1 conditional KO parasite line. Comparing the abundance of site-specific biotinylated peptides between wildtype and kinase deletion lines reveals the differential accessibility of proteins to biotinylation, indicating changes to localization, protein-protein interactions, or protein structure which are mediated by FIKK4.1 activity. We further show that FIKK4.1 is likely the only FIKK kinase that controls surface levels of PfEMP1, but not other surface antigens, on the infected red blood cell under standard culture conditions. We believe PerTurboID is broadly applicable to study the impact of genetic or environmental perturbation on a selected cellular niche.


Enzymes known as protein kinases regulate a huge variety of biological processes inside cells by attaching small tags known as phosphate groups onto specific locations on certain proteins. For example, the parasite that causes malaria infections in humans and great apes, injects a protein kinase called FIKK4.1 into certain cells in its host. This enzyme then adds phosphate groups to various parasite and host proteins that, in turn, causes them to form a large group of proteins (known as the cytoadhesion complex) to protect the parasite from being cleared by the hosts' immune defences. However, it remains unclear how and where the complex forms, and how the parasite regulates it. Proximity labelling is a well-established method that allows researchers to label and identify proteins that are near to a protein of interest. To investigate how the FIKK4.1 enzyme alters host cells to make the cytoadhesion complex, Davies et al. combined proximity labelling with methods that disturb the normal state of cells at a specific timepoint during development. The team used this new approach ­ named PerTurboID ­ to identify the proteins surrounding three components in the cytoadhesion complex. This made it possible to create a map of proteins that FIKK4.1 is likely to modify to build and control the cytoadhesion complex. Further experiments examined what happened to these surrounding proteins when FIKK4.1 was inactivated. This revealed that some protein targets of FIKK4.1 become either more or less accessible to other enzymes that attach a molecule known as biotin to proteins. This could be a result of structural changes in the cytoadhesion complex that are normally regulated by the FIKK4.1 kinase. In the future, PerTurboID may be useful to study how genetics or environmental changes affect other groups of proteins within specific environments inside cells, such as protein complexes required for DNA replication or cell division, or assembly of temporal structures required for cell movement.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Humanos , Parasitos/metabolismo , Proteínas de Protozoários/metabolismo , Plasmodium falciparum/metabolismo , Fosfotransferases/genética , Eritrócitos/parasitologia , Peptídeos/metabolismo , Malária Falciparum/parasitologia
9.
mBio ; 14(4): e0141323, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37489900

RESUMO

For its replication within red blood cells, the malaria parasite depends on a highly active and regulated lipid metabolism. Enzymes involved in lipid metabolic processes such as phospholipases are, therefore, potential drug targets. Here, using reverse genetics approaches, we show that only 1 out of the 19 putative phospholipases expressed in asexual blood stages of Plasmodium falciparum is essential for proliferation in vitro, pointing toward a high level of redundancy among members of this enzyme family. Using conditional mislocalization and gene disruption techniques, we show that this essential phosphoinositide-specific phospholipase C (PI-PLC, PF3D7_1013500) has a previously unrecognized essential role during intracellular parasite maturation, long before its previously perceived role in parasite egress and invasion. Subsequent lipidomic analysis suggests that PI-PLC mediates cleavage of phosphatidylinositol bisphosphate (PIP2) in schizont-stage parasites, underlining its critical role in regulating phosphoinositide levels in the parasite. IMPORTANCE The clinical symptoms of malaria arise due to repeated rounds of replication of Plasmodium parasites within red blood cells (RBCs). Central to this is an intense period of membrane biogenesis. Generation of membranes not only requires de novo synthesis and acquisition but also the degradation of phospholipids, a function that is performed by phospholipases. In this study, we investigate the essentiality of the 19 putative phospholipase enzymes that the human malaria parasite Plasmodium falciparum expresses during its replication within RBCs. We not only show that a high level of functional redundancy exists among these enzymes but, at the same time, also identify an essential role for the phosphoinositide-specific phospholipase C in parasite development and cleavage of the phospholipid phosphatidylinositol bisphosphate.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Humanos , Plasmodium falciparum/metabolismo , Parasitos/metabolismo , Fosfoinositídeo Fosfolipase C/metabolismo , Fosfolipases/genética , Fosfolipases/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Malária/metabolismo , Fosfolipídeos/metabolismo , Fosfatidilinositóis/metabolismo , Eritrócitos/parasitologia , Malária Falciparum/parasitologia
10.
Nat Commun ; 14(1): 2219, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37072430

RESUMO

Plasmodium falciparum causes the most severe form of malaria in humans. The protozoan parasite develops within erythrocytes to mature schizonts, that contain more than 16 merozoites, which egress and invade fresh erythrocytes. The aspartic protease plasmepsin X (PMX), processes proteins and proteases essential for merozoite egress from the schizont and invasion of the host erythrocyte, including the leading vaccine candidate PfRh5. PfRh5 is anchored to the merozoite surface through a 5-membered complex (PCRCR), consisting of Plasmodium thrombospondin-related apical merozoite protein, cysteine-rich small secreted protein, Rh5-interacting protein and cysteine-rich protective antigen. Here, we show that PCRCR is processed by PMX in micronemes to remove the N-terminal prodomain of PhRh5 and this activates the function of the complex unmasking a form that can bind basigin on the erythrocyte membrane and mediate merozoite invasion. The ability to activate PCRCR at a specific time in merozoite invasion most likely masks potential deleterious effects of its function until they are required. These results provide an important understanding of the essential role of PMX and the fine regulation of PCRCR function in P. falciparum biology.


Assuntos
Malária Falciparum , Plasmodium falciparum , Humanos , Animais , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Antígenos de Protozoários , Cisteína/metabolismo , Malária Falciparum/parasitologia , Eritrócitos/parasitologia , Merozoítos/metabolismo
11.
Int J Med Microbiol ; 313(3): 151579, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37030083

RESUMO

Plasmodium falciparum-related malaria represents a serious worldwide public health problem due to its high mortality rates. P. falciparum expresses rhoptry neck protein 4 (PfRON4) in merozoite and sporozoite rhoptries, it participates in tight junction-TJ formation via the AMA-1/RON complex and is refractory to complete genetic deletion. Despite this, which PfRON4 key regions interact with host cells remain unknown; such information would be useful for combating falciparum malaria. Thirty-two RON4 conserved region-derived peptides were chemically synthesised for determining and characterising PfRON4 regions having high host cell binding affinity (high activity binding peptides or HABPs). Receptor-ligand interaction/binding assays determined their specific binding capability, the nature of their receptors and their ability to inhibit in vitro parasite invasion. Peptides 42477, 42479, 42480, 42505 and 42513 had greater than 2% erythrocyte binding activity, whilst peptides 42477 and 42480 specifically bound to HepG2 membrane, both of them having micromolar and submicromolar range dissociation constants (Kd). Cell-peptide interaction was sensitive to treating erythrocytes with trypsin and/or chymotrypsin and HepG2 with heparinase I and chondroitinase ABC, suggesting protein-type (erythrocyte) and heparin and/or chondroitin sulphate proteoglycan receptors (HepG2) for PfRON4. Erythrocyte invasion inhibition assays confirmed HABPs' importance during merozoite invasion. PfRON4 800-819 (42477) and 860-879 (42480) regions specifically interacted with host cells, thereby supporting their inclusion in a subunit-based, multi-antigen, multistage anti-malarial vaccine.


Assuntos
Malária , Plasmodium falciparum , Animais , Humanos , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas de Transporte/metabolismo , Peptídeos , Eritrócitos/parasitologia , Ligação Proteica , Merozoítos/metabolismo , Hepatócitos/metabolismo , Antígenos de Protozoários
12.
Malar J ; 22(1): 5, 2023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-36604655

RESUMO

BACKGROUND: Polymorphisms in ATP2B4 coding for PMCA4b, the primary regulator of erythrocyte calcium concentration, have been shown by GWAS and cross-sectional studies to protect against severe malaria but the mechanism remains unknown. METHODS: Using a recall-by-genotype design, we investigated the impact of a common haplotype variant in ATP2B4 using in vitro assays that model erythrocyte stage malaria pathogenesis. Ninety-six donors representing homozygote (carriers of the minor allele, C/C), heterozygote (T/C) and wildtype (T/T) carriers of the tagging SNP rs1541252 were selected from a cohort of over 12,000 participants in the Keneba Biobank. RESULTS: Red blood cells (RBCs) from homozygotes showed reduced PMCA4b protein expression (mean fluorescence intensities (MFI = 2428 ± 124, 3544 ± 159 and 4261 ± 283], for homozygotes, heterozygotes and wildtypes respectively, p < 0.0001) and slower rates of calcium expulsion (calcium t½ ± SD = 4.7 ± 0.5, 1.8 ± 0.3 and 1.9 ± 0.4 min, p < 0.0001). Growth of a Plasmodium falciparum laboratory strain (FCR3) and two Gambian field isolates was decreased in RBCs from homozygotes compared to heterozygotes and wildtypes (p < 0.01). Genotype group did not affect parasite adhesion in vitro or var-gene expression in malaria-infected RBCs. Parasite growth was inhibited by a known inhibitor of PMCA4b, aurintricarboxylic acid (IC50 = 122uM CI: 110-134) confirming its sensitivity to calcium channel blockade. CONCLUSION: The data support the hypothesis that this ATP2B4 genotype, common in The Gambia and other malaria-endemic areas, protects against severe malaria through the suppression of parasitaemia during an infection. Reduction in parasite density plays a pivotal role in disease outcome by minimizing all aspects of malaria pathogenesis. Follow up studies are needed to further elucidate the mechanism of protection and to determine if this ATP2B4 genotype carries a fitness cost or increases susceptibility to other human disease.


Assuntos
Malária Falciparum , ATPases Transportadoras de Cálcio da Membrana Plasmática , Adulto , Humanos , Cálcio/metabolismo , Estudos Transversais , Eritrócitos/parasitologia , Gâmbia , Malária Falciparum/genética , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Plasmodium falciparum , Polimorfismo de Nucleotídeo Único
13.
Microb Pathog ; 175: 105954, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36574865

RESUMO

BACKGROUND: Babesiosis is an emerging zoonosis worldwide that is caused by tick-borne apicomplexans, Babesia spp., which threatens the health of domesticated and wild mammals and even humans. Although it has done serious harm to animal husbandry and public health, the study of Babesia is still progressing slowly. Until now, no effective anti-Babesia vaccines have been available, and administration of combined drugs tends to produce side effects. Therefore, non-targeted metabolomics was employed in the present study to examine the temporal dynamic changes in the metabolic profile of the infected erythrocytes. The goal was to obtain new insight into pathogenesis of Babesia and to explore vaccine candidates or novel drug targets. METHODS: C57BL/6 mice were infected with B. microti and erythrocytes at different time points (0, 3, 6 , 9, 12, and 22-days post-infection) were subjected to parasitemia surveillance and then metabolomics analysis using liquid chromatography-mass spectrometry (LC-MS). Multivariate statistical analyses were performed to clearly separate and identify dysregulated metabolites in Babesia-infected mice. The analyses included principal components analysis (PCA) and orthogonal partial least squares-discrimination analysis (OPLS-DA). The time-series trends of the impacted molecules were analyzed using the R package Mfuzz and the fuzzy clustering principle. The temporal profiling of amino acids, lipids, and nucleotides in blood cells infected with B. microti were also investigated. RESULTS: B. microti infection resulted in a fast increase of parasitemia and serious alteration of the mouse metabolites. Through LC-MS metabolomics analysis, 10,289 substance peaks were detected and annotated to 3,705 components during the analysis period. There were 1,166 dysregulated metabolites, which were classified into 8 clusters according to the temporal trends. Consistent with the trend of parasitemia, the numbers of differential metabolites reached a peak of 525 at 6-days post-infection (dpi). Moreover, the central carbon metabolism in cancer demonstrated the most serious change during the infection process except for that observed at 6 dpi. Sabotage occurred in components involved in the TCA cycle, amino acids, lipids, and nucleotide metabolism. CONCLUSION: Our findings revealed a great alteration in the metabolites of Babesia-infected mice and shed new light on the pathogenesis of B. microti at the metabolic level. The results might lead to novel information about the mechanisms of pathopoiesis, babesisosis, and anti-parasite drug/vaccine development in the future.


Assuntos
Babesia microti , Humanos , Animais , Camundongos , Parasitemia , Camundongos Endogâmicos C57BL , Eritrócitos/parasitologia , Lipídeos , Mamíferos
14.
Front Cell Infect Microbiol ; 12: 1060202, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36530423

RESUMO

The cation efflux pump Plasmodium falciparum ATPase 4 (PfATP4) maintains Na+ homeostasis in malaria parasites and has been implicated in the mechanism of action of many structurally diverse antimalarial agents, including >7% of the antimalarial compounds in the Medicines for Malaria Venture's 'Malaria Box' and 'Pathogen Box'. Recent screens of the 'Malaria Box' and 'Pathogen Box' revealed that many PfATP4 inhibitors prevent parasites from exiting their host red blood cell (egress) or entering new host cells (invasion), suggesting that these compounds may have additional molecular targets involved in egress or invasion. Here, we demonstrate that five PfATP4 inhibitors reduce egress but not invasion. These compounds appear to inhibit egress by blocking the activation of protein kinase G, an enzyme that, once stimulated, rapidly activates parasite egress. We establish a direct link between egress and PfATP4 function by showing that the inhibition of egress is attenuated in a Na+-depleted environment and in parasites with a mutation in pfatp4. Finally, we show that PfATP4 inhibitors induce host cell lysis when administered prior to the completion of parasite replication. Since host cell lysis mimics egress but is not followed by invasion, this phenomenon likely explains why several PfATP4 inhibitors were previously classified as invasion inhibitors. Collectively, our results confirm that PfATP4-mediated Na+ efflux is critical to the regulation of parasite egress.


Assuntos
Antimaláricos , Proteínas de Transporte de Cátions , Malária Falciparum , Malária , Animais , Humanos , Plasmodium falciparum/genética , Esquizontes/metabolismo , Adenosina Trifosfatases/genética , Antimaláricos/farmacologia , Malária Falciparum/parasitologia , Eritrócitos/parasitologia , Malária/metabolismo , Proteínas de Transporte de Cátions/genética , Sódio/metabolismo , Sódio/uso terapêutico , Íons/metabolismo
15.
mBio ; 13(6): e0309622, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36445080

RESUMO

During vertebrate infection, obligate intracellular malaria parasites develop within a parasitophorous vacuole, which constitutes the interface between the parasite and its hepatocyte or erythrocyte host cells. To traverse this barrier, Plasmodium spp. utilize a dual-function pore formed by EXP2 for nutrient transport and, in the context of the PTEX translocon, effector protein export across the vacuole membrane. While critical to blood-stage survival, less is known about EXP2/PTEX function in the liver stage, although major differences in the export mechanism are suggested by absence of the PTEX unfoldase HSP101 in the intrahepatic vacuole. Here, we employed the glucosamine-activated glmS ribozyme to study the role of EXP2 during Plasmodium berghei liver-stage development in hepatoma cells. Insertion of the glmS sequence into the exp2 3' untranslated region (UTR) enabled glucosamine-dependent depletion of EXP2 after hepatocyte invasion, allowing separation of EXP2 function during intrahepatic development from a recently reported role in hepatocyte invasion. Postinvasion EXP2 knockdown reduced parasite size and largely abolished expression of the mid- to late-liver-stage marker LISP2. As an orthogonal approach to monitor development, EXP2-glmS parasites and controls were engineered to express nanoluciferase. Activation of glmS after invasion substantially decreased luminescence in hepatoma monolayers and in culture supernatants at later time points corresponding to merosome detachment, which marks the culmination of liver-stage development. Collectively, our findings extend the utility of the glmS ribozyme to study protein function in the liver stage and reveal that EXP2 is important for intrahepatic parasite development, indicating that PTEX components also function at the hepatocyte-parasite interface. IMPORTANCE After the mosquito bite that initiates a Plasmodium infection, parasites first travel to the liver and develop in hepatocytes. This liver stage is asymptomatic but necessary for the parasite to transition to the merozoite form, which infects red blood cells and causes malaria. To take over their host cells, avoid immune defenses, and fuel their growth, these obligately intracellular parasites must import nutrients and export effector proteins across a vacuole membrane in which they reside. In the blood stage, these processes depend on a translocon called PTEX, but it is unclear if PTEX also functions during the liver stage. Here, we adapted the glmS ribozyme to control expression of EXP2, the membrane pore component of PTEX, during the liver stage of the rodent malaria parasite Plasmodium berghei. Our results show that EXP2 is important for intracellular development in the hepatocyte, revealing that PTEX components are also functionally important during liver-stage infection.


Assuntos
Eritrócitos , Hepatócitos , Malária , Plasmodium berghei , Proteínas de Protozoários , Carcinoma Hepatocelular , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Neoplasias Hepáticas , Malária/genética , Malária/metabolismo , Malária/parasitologia , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/genética , Transporte Proteico/genética , Transporte Proteico/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , RNA Catalítico/metabolismo , Animais , Camundongos , Hepatócitos/metabolismo , Hepatócitos/parasitologia
16.
Curr Opin Microbiol ; 70: 102221, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36242898

RESUMO

Targeting critical host factors is an emerging concept in the treatment of infectious diseases. As obligate pathogens of erythrocytes, the Plasmodium spp. parasites that cause malaria must exploit erythroid host factors for their survival. However, our understanding of this important aspect of the malaria lifecycle is limited, in part because erythrocytes are enucleated cells that lack a nucleus and DNA, rendering them genetically intractable. Recent advances in genetic analysis of the erythroid lineage using small-hairpin RNAs and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated nuclease 9 (Cas9) in red-blood cells derived from stem cells have generated new insights into the functions of several candidate host factors for Plasmodium parasites. Along with efforts in other hematopoietic cells, these advances have also laid a strong foundation for genetic screens to identify novel erythrocyte host factors for malaria.


Assuntos
Malária , Plasmodium , Humanos , Linhagem da Célula , Eritrócitos/parasitologia , Plasmodium falciparum/genética , Plasmodium/genética , Malária/parasitologia , Sistemas CRISPR-Cas
17.
Nat Commun ; 13(1): 5746, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36180431

RESUMO

Diverse compounds target the Plasmodium falciparum Na+ pump PfATP4, with cipargamin and (+)-SJ733 the most clinically-advanced. In a recent clinical trial for cipargamin, recrudescent parasites emerged, with most having a G358S mutation in PfATP4. Here, we show that PfATP4G358S parasites can withstand micromolar concentrations of cipargamin and (+)-SJ733, while remaining susceptible to antimalarials that do not target PfATP4. The G358S mutation in PfATP4, and the equivalent mutation in Toxoplasma gondii ATP4, decrease the sensitivity of ATP4 to inhibition by cipargamin and (+)-SJ733, thereby protecting parasites from disruption of Na+ regulation. The G358S mutation reduces the affinity of PfATP4 for Na+ and is associated with an increase in the parasite's resting cytosolic [Na+]. However, no defect in parasite growth or transmissibility is observed. Our findings suggest that PfATP4 inhibitors in clinical development should be tested against PfATP4G358S parasites, and that their combination with unrelated antimalarials may mitigate against resistance development.


Assuntos
Antimaláricos , Malária Falciparum , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , ATPases Transportadoras de Cálcio , Eritrócitos/parasitologia , Humanos , Indóis , Íons , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Mutação , Plasmodium falciparum , Sódio , Compostos de Espiro
18.
mBio ; 13(4): e0163522, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35862778

RESUMO

In model organisms, type IV ATPases (P4-ATPases) require cell division control protein 50 (CDC50) chaperones for their phospholipid flipping activity. In the malaria parasite Plasmodium falciparum, guanylyl cyclase alpha (GCα) is an integral membrane protein that is essential for release (egress) of merozoites from their host erythrocytes. GCα is unusual in that it contains both a C-terminal cyclase domain and an N-terminal P4-ATPase domain of unknown function. We sought to investigate whether any of the three CDC50 orthologues (termed A, B, and C) encoded by P. falciparum are required for GCα function. Using gene tagging and conditional gene disruption, we demonstrate that CDC50B and CDC50C but not CDC50A are expressed in the clinically important asexual blood stages and that CDC50B is a binding partner of GCα whereas CDC50C is the binding partner of another putative P4-ATPase, phospholipid-transporting ATPase 2 (ATP2). Our findings indicate that CDC50B has no essential role for intraerythrocytic parasite maturation but modulates the rate of parasite egress by interacting with GCα for optimal cGMP synthesis. In contrast, CDC50C is essential for blood stage trophozoite maturation. Additionally, we find that the CDC50C-ATP2 complex may influence parasite endocytosis of host cell hemoglobin and consequently hemozoin formation. IMPORTANCE Malaria morbidity arises due to successive rounds of replication of Plasmodium parasites within red blood cells. Mature daughter merozoites are released from infected erythrocytes to invade new cells in a tightly regulated process termed egress. Previous studies have shown that a unique bifunctional guanylyl cyclase, GCα, initiates egress by synthesis of cGMP. GCα has an N-terminal P4-ATPase domain of unknown function. In model organisms, P4-ATPases function through interaction with a CDC50 partner protein. Here, we investigate the role of CDC50 orthologues in P. falciparum and show that GCα binds CDC50B, an interaction that regulates egress efficiency. We also find that CDC50C is essential and binds a putative P4-ATPase, ATP2, in a complex that influences endocytosis of host hemaglobin. Our results highlight the heterogenous and critical role of CDC50 proteins in P. falciparum.


Assuntos
Malária Falciparum , Malária , Adenosina Trifosfatases/genética , Animais , Eritrócitos/parasitologia , Guanilato Ciclase , Humanos , Malária Falciparum/parasitologia , Merozoítos/fisiologia , Fosfolipídeos , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Trofozoítos/metabolismo
19.
Methods Mol Biol ; 2470: 659-672, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35881381

RESUMO

Apoptosis is conventionally regarded as an evolutionarily conserved and genetically controlled process of programmed cell death confined to metazoan organisms. However, recently, conserved features of apoptosis have also been demonstrated in unicellular eukaryotes (Holzmuller et al. Parasitology 132:S19-S32, 2006; Le Chat et al. Mol Biochem Parasitol 153:41-47, 2007; Madeo et al. Curr Opin Microbiol 7:655-660, 2004; Welburn et al. Parasitology 132:S7-S18, 2006; Jensen et al. Science 216:1230-1233, 1982) including malaria parasites (Al-Olayan et al. Int J Parasitol 32:1133-1143, 2002; Ch'ng et al. Cell Death Dis 1:e26, 2010; Meslin et al. J Infect Dis 195:1852-1859, 2007; Picot et al. Trans R Soc Trop Med Hyg 91:590-591, 1997; Raj et al. Nature 582:104-108, 2020). P. falciparum glutamic-acid-rich protein (PfGARP) is an antigen of 80 kDa that is uniquely expressed on the exofacial surface of red blood cells (RBCs) infected by early-to-late-trophozoite-stage P. falciparum parasites (Raj et al. Nature 582:104-108, 2020). We have recently demonstrated that antibodies against PfGARP bind to the PfGARP displayed on the surface of P. falciparum trophozoite-infected RBCs and trigger apoptosis in the intracellular parasites (Raj et al. Nature 582:104-108, 2020). This is the first demonstration of antibody-induced apoptosis in blood-stage malaria parasites and is characterized by several conserved features such as crisis form morphology, loss of mitochondrial membrane potential, loss of integrity of food vacuole, activation of caspase-like cysteine proteases, and fragmentation of chromosomal DNA. Here we describe the assays used to detect these features of apoptosis in the mature blood stage of malaria parasites.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Apoptose , Eritrócitos/parasitologia , Humanos , Malária/parasitologia , Malária Falciparum/parasitologia , Plasmodium falciparum/genética
20.
Nat Commun ; 13(1): 3307, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35676275

RESUMO

Severe falciparum malaria is a major cause of preventable child mortality in sub-Saharan Africa. Plasma concentrations of P. falciparum Histidine-Rich Protein 2 (PfHRP2) have diagnostic and prognostic value in severe malaria. We investigate the potential use of plasma PfHRP2 and the sequestration index (the ratio of PfHRP2 to parasite density) as quantitative traits for case-only genetic association studies of severe malaria. Data from 2198 Kenyan children diagnosed with severe malaria, genotyped for 14 major candidate genes, show that polymorphisms in four major red cell genes that lead to hemoglobin S, O blood group, α-thalassemia, and the Dantu blood group, are associated with substantially lower admission plasma PfHRP2 concentrations, consistent with protective effects against extensive parasitized erythrocyte sequestration. In contrast the known protective ATP2B4 polymorphism is associated with higher plasma PfHRP2 concentrations, lower parasite densities and a higher sequestration index. We provide testable hypotheses for the mechanism of protection of ATP2B4.


Assuntos
Antígenos de Grupos Sanguíneos , Eritrócitos , Malária Falciparum , Antígenos de Protozoários/genética , Antígenos de Protozoários/metabolismo , Biomassa , Antígenos de Grupos Sanguíneos/metabolismo , Criança , Eritrócitos/parasitologia , Humanos , Quênia , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA