Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.663
Filter
1.
PLoS One ; 19(8): e0304842, 2024.
Article in English | MEDLINE | ID: mdl-39116045

ABSTRACT

The retromer is a cellular structure that recruits and recycles proteins inside the cell. In mammalian and yeast, the retromer components have been widely studied, but very little in parasites. In yeast, it is formed by a SNX-BAR membrane remodeling heterodimer and the cargo selecting complex (CSC), composed by three proteins. One of them, the Vps26 protein, possesses a flexible and intrinsically disordered region (IDR), that facilitates interactions with other proteins and contributes to the retromer binding to the endosomal membrane. In Entamoeba histolytica, the protozoan parasite responsible for human amoebiasis, the retromer actively participates during the high mobility and phagocytosis of trophozoites, but the molecular details in these events, are almost unknown. Here, we studied the EhVps26 role in phagocytosis. Bioinformatic analyses of EhVps26 revealed a typical arrestin folding structure of the protein, and a long and charged IDR, as described in other systems. EhVps26 molecular dynamics simulations (MDS) allowed us to predict binding pockets for EhVps35, EhSNX3, and a PX domain-containing protein; these pockets were disorganized in a EhVps26 truncated version lacking the IDR. The AlphaFold2 software predicted the interaction of EhVps26 with EhVps35, EhVps29 and EhSNX3, in a model similar to the reported mammalian crystals. By confocal and transmission electron microscopy, EhVps26 was found in the trophozoites plasma membrane, cytosol, endosomes, and Golgi-like apparatus. During phagocytosis, it followed the erythrocytes pathway, probably participating in cargoes selection and recycling. Ehvps26 gene knocking down evidenced that the EhVps26 protein is necessary for efficient phagocytosis.


Subject(s)
Computational Biology , Entamoeba histolytica , Phagocytosis , Protozoan Proteins , Entamoeba histolytica/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Computational Biology/methods , Humans , Molecular Dynamics Simulation , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/chemistry , Protein Binding , Amino Acid Sequence , Erythrocytes/parasitology , Erythrocytes/metabolism
2.
Trop Biomed ; 41(2): 190-195, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-39154272

ABSTRACT

The Plasmodium secreted protein with an altered thrombospondin repeat (SPATR) has been known to play an important role in the malaria parasite's invasion into host erythrocytes. This protein is immunogenic and has been considered as one of the potential vaccine candidates against malaria parasite infection. Thus far, only a handful immunological studies have been carried out on P. knowlesi SPATR (PkSPATR), and none of these studies investigated the immunoprotective properties of the protein. In the present study, the ability of anti-PkSPATR antibodies to inhibit invasion of human erythrocytes was assessed in an in vitro merozoite invasion inhibition assay. The antibodies were harvested from the serum of a rabbit which was immunised with recombinat PkSPATR. Results from the merozoite invasion inhibition assay revealed significant antibody invasion inhibitory activity in a concentration dependent manner (concentration range: 0.375 - 3.00 mg/ml) with inhibition rate ranging from 20% to 32%. Future studies, such as anti-PkSPATR antibodies inhibitory effect on sporozoite invasion of human liver cells, need to be carried out to assess the potential of PkSPATR as a knowlesi malaria vaccine candidate.


Subject(s)
Antibodies, Protozoan , Erythrocytes , Merozoites , Plasmodium knowlesi , Protozoan Proteins , Plasmodium knowlesi/immunology , Humans , Erythrocytes/parasitology , Rabbits , Animals , Antibodies, Protozoan/immunology , Protozoan Proteins/immunology , Merozoites/immunology , Thrombospondins/immunology , Malaria Vaccines/immunology
3.
FASEB J ; 38(15): e23856, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39092913

ABSTRACT

Merozoites utilize sialic acids on the red blood cell (RBC) cell surface to rapidly adhere to and invade the RBCs. Newcastle disease virus (NDV) displays a strong affinity toward membrane-bound sialic acids. Incubation of NDV with the malaria parasites dose-dependently reduces its cellular viability. The antiplasmodial activity of NDV is specific, as incubation with Japanese encephalitis virus, duck enteritis virus, infectious bronchitis virus, and influenza virus did not affect the parasite propagation. Interestingly, NDV is reducing more than 80% invasion when RBCs are pretreated with the virus. Removal of the RBC surface proteins or the NDV coat proteins results in disruption of the virus binding to RBC. It suggests the involvement of specific protein: ligand interaction in virus binding. We established that the virus engages with the parasitized RBCs (PRBCs) through its hemagglutinin neuraminidase (HN) protein by recognizing sialic acid-containing glycoproteins on the cell surface. Blocking of the HN protein with free sialic acid or anti-HN antibodies abolished the virus binding as well as its ability to reduce parasite growth. Interestingly, the purified HN from the virus alone could inhibit the parasite's growth in a dose-dependent manner. NDV binds strongly to knobless murine parasite strain Plasmodium yoelii and restricted the parasite growth in mice. Furthermore, the virus was found to preferentially target the PRBCs compared to normal erythrocytes. Immunolocalization studies reveal that NDV is localized on the plasma membrane as well as weakly inside the PRBC. NDV causes neither any infection nor aggregation of the human RBCs. Our findings suggest that NDV is a potential candidate for developing targeted drug delivery platforms for the Plasmodium-infected RBCs.


Subject(s)
Erythrocytes , N-Acetylneuraminic Acid , Newcastle disease virus , Newcastle disease virus/physiology , Newcastle disease virus/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism , Animals , N-Acetylneuraminic Acid/metabolism , Humans , Plasmodium yoelii/metabolism , Mice , HN Protein/metabolism , Malaria/parasitology , Malaria/metabolism
4.
Malar J ; 23(1): 227, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090669

ABSTRACT

BACKGROUND: Plasmodium falciparum, the malaria-causing parasite, is a leading cause of infection-induced deaths worldwide. The preferred treatment approach is artemisinin-based combination therapy, which couples fast-acting artemisinin derivatives with longer-acting drugs, such as lumefantrine, mefloquine, and amodiaquine. However, the urgency for new treatments has risen due to the parasite's growing resistance to existing therapies. In this study, a common characteristic of the P. falciparum proteome-stretches of poly-lysine residues, such as those found in proteins related to adhesion and pathogenicity-is investigated for its potential to treat infected erythrocytes. METHODS: This study utilizes in vitro culturing of intra-erythrocytic P. falciparum to assess the ability of poly-lysine peptides to inhibit the parasite's growth, measured via flow cytometry of acridine orange-stained infected erythrocytes. The inhibitory effect of many poly-lysine lengths and modifications were tested this way. Affinity pull-downs and mass spectrometry were performed to identify the proteins interacting with these poly-lysines. RESULTS: A single dose of these poly-basic peptides can successfully diminish parasitemia in human erythrocytes in vitro with minimal toxicity. The effectiveness of the treatment correlates with the length of the poly-lysine peptide, with 30 lysine peptides supporting the eradication of erythrocytic parasites within 72 h. PEG-ylation of the poly-lysine peptides or utilizing poly-lysine dendrimers and polymers retains or increases parasite clearance efficiency and bolsters the stability of these potential new therapeutics. Lastly, affinity pull-downs and mass-spectrometry identify P. falciparum's outer membrane proteins as likely targets for polybasic peptide medications. CONCLUSION: Since poly-lysine dendrimers are already FDA-approved for drug delivery and this study displays their potency against intraerythrocytic P. falciparum, their adaptation as anti-malarial drugs presents a promising new therapeutic strategy for malaria.


Subject(s)
Antimalarials , Erythrocytes , Plasmodium falciparum , Plasmodium falciparum/drug effects , Antimalarials/pharmacology , Antimalarials/chemistry , Erythrocytes/drug effects , Erythrocytes/parasitology , Peptides/pharmacology , Peptides/chemistry , Humans , Polymers/pharmacology , Polymers/chemistry , Polylysine/pharmacology , Polylysine/chemistry
5.
Br J Haematol ; 205(2): 414-415, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38977858

ABSTRACT

For over a century, the need to identify malaria in the peripheral blood has been the driving force behind the development of fundamental clinical microscopy techniques. In the study by Moysis et al., artificial intelligence-based model was utilized to identify and provide quantitative morphological characteristics of red blood cells typical to severe malaria anaemia, irrespective to the actual presence of visible parasites. Commentary on: Moysis et al. Leveraging deep learning for detecting red blood cell morphological changes in blood films from children with severe malaria anaemia. Br J Haematol 2024;205:699-710.


Subject(s)
Artificial Intelligence , Erythrocytes , Malaria , Humans , Malaria/blood , Malaria/diagnosis , Erythrocytes/parasitology , Anemia/blood , Anemia/diagnosis , Child
6.
J Cell Sci ; 137(20)2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38962997

ABSTRACT

Lipid droplets (LDs) are organelles that are central to lipid and energy homeostasis across all eukaryotes. In the malaria-causing parasite Plasmodium falciparum the roles of LDs in lipid acquisition from its host cells and their metabolism are poorly understood, despite the high demand for lipids in parasite membrane synthesis. We systematically characterised LD size, composition and dynamics across the disease-causing blood infection. Applying split fluorescence emission analysis and three-dimensional (3D) focused ion beam-scanning electron microscopy (FIB-SEM), we observed a decrease in LD size in late schizont stages. LD contraction likely signifies a switch from lipid accumulation to lipid utilisation in preparation for parasite egress from host red blood cells. We demonstrate connections between LDs and several parasite organelles, pointing to potential functional interactions. Chemical inhibition of triacylglyerol (TAG) synthesis or breakdown revealed essential LD functions for schizogony and in counteracting lipid toxicity. The dynamics of lipid synthesis, storage and utilisation in P. falciparum LDs might provide a target for new anti-malarial intervention strategies.


Subject(s)
Erythrocytes , Lipid Droplets , Malaria, Falciparum , Plasmodium falciparum , Plasmodium falciparum/metabolism , Lipid Droplets/metabolism , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism , Lipid Metabolism , Triglycerides/metabolism
7.
Biochim Biophys Acta Gen Subj ; 1868(9): 130665, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38969256

ABSTRACT

BACKGROUND: The malaria parasite Plasmodium falciparum replicates within red blood cells, then ruptures the cell in a process called egress in order to continue its life cycle. Egress is regulated by a proteolytic cascade involving an essential parasite subtilisin-like serine protease called SUB1. Maturation of SUB1 initiates in the parasite endoplasmic reticulum with autocatalytic cleavage of an N-terminal prodomain (p31), which initially remains non-covalently bound to the catalytic domain, p54. Further trafficking of the p31-p54 complex results in formation of a terminal p47 form of the SUB1 catalytic domain. Recent work has implicated a parasite aspartic protease, plasmepsin X (PMX), in maturation of the SUB1 p31-p54 complex through controlled cleavage of the prodomain p31. METHODS: Here we use biochemical and enzymatic analysis to examine the activation of SUB1 by PMX. RESULTS: We show that both p31 and p31-p54 are largely dimeric under the relatively acidic conditions to which they are likely exposed to PMX in the parasite. We confirm the sites within p31 that are cleaved by PMX and determine the order of cleavage. We find that cleavage by PMX results in rapid loss of the capacity of p31 to act as an inhibitor of SUB1 catalytic activity and we directly demonstrate that exposure to PMX of recombinant p31-p54 complex activates SUB1 activity. CONCLUSIONS: Our results confirm that precise, PMX-mediated cleavage of the SUB1 prodomain activates SUB1 enzyme activity. GENERAL SIGNIFICANCE: Our findings elucidate the role of PMX in activation of SUB1, a key effector of malaria parasite egress.


Subject(s)
Aspartic Acid Endopeptidases , Plasmodium falciparum , Protozoan Proteins , Plasmodium falciparum/enzymology , Plasmodium falciparum/metabolism , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Proteolysis , Humans , Subtilisins/metabolism , Catalytic Domain , Protein Domains , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism
8.
J Infect Dis ; 230(1): 263-270, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052743

ABSTRACT

Pathogens such as Plasmodium, Babesia, and Theileria invade and multiply within host red blood cells, leading to the pathological consequences of malaria, babesiosis, and theileriosis. Establishing continuous in vitro culture systems and suitable animal models is crucial for studying these pathogens. This review spotlights the Babesia duncani in culture-in mouse (ICIM) model as a promising resource for advancing research on the biology, pathogenicity, and virulence of intraerythrocytic parasites. The model offers practical benefits, encompassing well-defined culture conditions, ease of manipulation, and a well-annotated genome. Moreover, B. duncani serves as a surrogate system for drug discovery, facilitating the evaluation of new antiparasitic drugs in vitro and in animals, elucidating their modes of action, and uncovering potential resistance mechanisms. The B. duncani ICIM model thus emerges as a multifaceted tool with profound implications, promising advancements in our understanding of parasitic biology and shaping the development of future therapies.


Subject(s)
Babesia , Babesiosis , Disease Models, Animal , Erythrocytes , Animals , Babesia/drug effects , Babesia/genetics , Erythrocytes/parasitology , Mice , Babesiosis/drug therapy , Babesiosis/parasitology , Antiparasitic Agents/therapeutic use , Antiparasitic Agents/pharmacology , Humans , Virulence
9.
Biomolecules ; 14(7)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39062495

ABSTRACT

Parasite-derived new permeation pathways (NPPs) expressed at the red blood cell (RBC) membrane enable Plasmodium parasites to take up nutrients from the plasma to facilitate their survival. Thus, NPPs represent a potential novel therapeutic target for malaria. The putative channel component of the NPP in the human malaria parasite P. falciparum is encoded by mutually exclusively expressed clag3.1/3.2 genes. Complicating the study of the essentiality of these genes to the NPP is the addition of three clag paralogs whose contribution to the P. falciparum channel is uncertain. Rodent malaria P. berghei contains only two clag genes, and thus studies of P. berghei clag genes could significantly aid in dissecting their overall contribution to NPP activity. Previous methods for determining NPP activity in a rodent model have utilised flux-based assays of radioisotope-labelled substrates or patch clamping. This study aimed to ratify a streamlined haemolysis assay capable of assessing the functionality of P. berghei NPPs. Several isotonic lysis solutions were tested for their ability to preferentially lyse infected RBCs (iRBCs), leaving uninfected RBCs (uRBCs) intact. The osmotic lysis assay was optimised and validated in the presence of NPP inhibitors to demonstrate the uptake of the lysis solution via the NPPs. Guanidinium chloride proved to be the most efficient reagent to use in an osmotic lysis assay to establish NPP functionality. Furthermore, following treatment with guanidinium chloride, ring-stage parasites could develop into trophozoites and schizonts, potentially enabling use of guanidinium chloride for parasite synchronisation. This haemolysis assay will be useful for further investigation of NPPs in P. berghei and could assist in validating its protein constituents.


Subject(s)
Erythrocytes , Guanidine , Hemolysis , Malaria , Plasmodium berghei , Plasmodium berghei/drug effects , Animals , Hemolysis/drug effects , Guanidine/pharmacology , Erythrocytes/parasitology , Erythrocytes/metabolism , Erythrocytes/drug effects , Mice , Malaria/drug therapy , Malaria/parasitology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Humans
10.
Evolution ; 78(8): 1513-1514, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38887989

ABSTRACT

The virulence of parasites is expected to reflect an evolutionary tradeoff between increasing proliferation rates that enhance transmission and host mortality which curtails transmission. However, host resource availability may also limit parasites' proliferation rate. To understand the role of resource limitation as a driver of virulence evolution, Pak et al. (2024) use a within-host model of red blood cell (RBC) invasion by Plasmodium chabaudi. They find that within-host resource consumption limits the evolution of the parasite's proliferation rate, as the depletion of RBCs during infection results in intermediate optimal virulence. These results suggest that resource limitation, rather than host mortality, may drive the evolution of virulence.


Subject(s)
Biological Evolution , Plasmodium chabaudi , Virulence , Plasmodium chabaudi/genetics , Plasmodium chabaudi/pathogenicity , Erythrocytes/parasitology , Host-Parasite Interactions , Animals , Malaria/parasitology
11.
Infect Immun ; 92(7): e0021524, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38884473

ABSTRACT

Human babesiosis is a malaria-like illness caused by protozoan parasites of the genus Babesia. Babesia microti is responsible for most cases of human babesiosis in the United States, particularly in the Northeast and the Upper Midwest. Babesia microti is primarily transmitted to humans through the bite of infected deer ticks but also through the transfusion of blood components, particularly red blood cells. There is a high risk of severe and even fatal disease in immunocompromised patients. To date, serology testing relies on an indirect immunofluorescence assay that uses the whole Babesia microti antigen. Here, we report the construction of phage display cDNA libraries from Babesia microti-infected erythrocytes as well as human reticulocytes obtained from donors with hereditary hemochromatosis. Plasma samples were obtained from patients who were or had been infected with Babesia microti. The non-specific antibody reactivity of these plasma samples was minimized by pre-exposure to the human reticulocyte library. Using this novel experimental strategy, immunoreactive segments were identified in three Babesia microti antigens termed BmSA1 (also called BMN1-9; BmGPI12), BMN1-20 (BMN1-17; Bm32), and BM4.12 (N1-15). Moreover, our findings indicate that the major immunoreactive segment of BmSA1 does not overlap with the segment that mediates BmSA1 binding to mature erythrocytes. When used in combination, the three immunoreactive segments form the basis of a sensitive and comprehensive diagnostic immunoassay for human babesiosis, with implications for vaccine development.


Subject(s)
Antigens, Protozoan , Babesia microti , Babesiosis , Gene Library , Babesia microti/immunology , Babesia microti/genetics , Humans , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Babesiosis/immunology , Babesiosis/parasitology , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Erythrocytes/parasitology , Erythrocytes/immunology , Cell Surface Display Techniques , Animals
12.
Infect Immun ; 92(7): e0048123, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38837339

ABSTRACT

The currently accepted initiation of Babesia infection describes a sporozoite stage infused into the host, along with other saliva components, by the tick vector. This sporozoite can enter and initiate erythrocyte infection directly. In the particular case of Babesia microti, however, that sporozoite loses the ability to further propagate in vitro once deprived of its natural host. True B. sensu stricto do not require the host collaboration described in this study. Hence it has become a current topic of research involving B. microti (B. sensu lato), a rather unique species that requires host collaboration to maintain an erythrocyte propagation cycle. The main attachment protein is synthesized by this parasite in excess and exported to the host from the erythrocyte infrastructure to immunize the host at all stages of infection. The synthesis of host immune IgM antibody is necessary for the propagation of B. microti, being central to entry into uninfected host erythrocytes. Sequential use of the host immune system then involves complement factor C3b to complete the three-part assembly necessary to initiate the rhoptry sequence for invasion of uninfected erythrocytes and further propagation. These several components must be furnished within the in vitro culture medium and the sequence of these reactions is discussed. The corollary view of the parasite survival versus the host immune defenses is also discussed as it involves the same host factors promoting continuing parasite growth. This is the first description of continuous in vitro propagation of B. microti.


Subject(s)
Babesia microti , Erythrocytes , Animals , Humans , Babesia microti/immunology , Babesiosis/parasitology , Babesiosis/immunology , Erythrocytes/parasitology , Host-Parasite Interactions
13.
PLoS Pathog ; 20(6): e1012360, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38935780

ABSTRACT

The cGMP-dependent protein kinase (PKG) is the sole cGMP sensor in malaria parasites, acting as an essential signalling hub to govern key developmental processes throughout the parasite life cycle. Despite the importance of PKG in the clinically relevant asexual blood stages, many aspects of malarial PKG regulation, including the importance of phosphorylation, remain poorly understood. Here we use genetic and biochemical approaches to show that reduced cGMP binding to cyclic nucleotide binding domain B does not affect in vitro kinase activity but prevents parasite egress. Similarly, we show that phosphorylation of a key threonine residue (T695) in the activation loop is dispensable for kinase activity in vitro but is essential for in vivo PKG function, with loss of T695 phosphorylation leading to aberrant phosphorylation events across the parasite proteome and changes to the substrate specificity of PKG. Our findings indicate that Plasmodium PKG is uniquely regulated to transduce signals crucial for malaria parasite development.


Subject(s)
Cyclic GMP-Dependent Protein Kinases , Cyclic GMP , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Phosphorylation , Cyclic GMP/metabolism , Malaria/parasitology , Malaria/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Animals , Plasmodium falciparum/metabolism , Plasmodium falciparum/genetics , Humans , Signal Transduction , Erythrocytes/parasitology , Erythrocytes/metabolism
14.
Immunohorizons ; 8(6): 442-456, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38916585

ABSTRACT

Malaria is a serious vector-borne disease characterized by periodic episodes of high fever and strong immune responses that are coordinated with the daily synchronized parasite replication cycle inside RBCs. As immune cells harbor an autonomous circadian clock that controls various aspects of the immune response, we sought to determine whether the intensity of the immune response to Plasmodium spp., the parasite causing malaria, depends on time of infection. To do this, we developed a culture model in which mouse bone marrow-derived macrophages are stimulated with RBCs infected with Plasmodium berghei ANKA (iRBCs). Lysed iRBCs, but not intact iRBCs or uninfected RBCs, triggered an inflammatory immune response in bone marrow-derived macrophages. By stimulating at four different circadian time points (16, 22, 28, or 34 h postsynchronization of the cells' clock), 24-h rhythms in reactive oxygen species and cytokines/chemokines were found. Furthermore, the analysis of the macrophage proteome and phosphoproteome revealed global changes in response to iRBCs that varied according to circadian time. This included many proteins and signaling pathways known to be involved in the response to Plasmodium infection. In summary, our findings show that the circadian clock within macrophages determines the magnitude of the inflammatory response upon stimulation with ruptured iRBCs, along with changes of the cell proteome and phosphoproteome.


Subject(s)
Circadian Rhythm , Erythrocytes , Macrophages , Malaria , Plasmodium berghei , Animals , Macrophages/immunology , Macrophages/parasitology , Macrophages/metabolism , Mice , Erythrocytes/parasitology , Erythrocytes/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/immunology , Circadian Rhythm/immunology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Cytokines/metabolism , Circadian Clocks/immunology , Cells, Cultured , Proteome/metabolism
15.
Microbiol Spectr ; 12(7): e0072724, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38864616

ABSTRACT

A hallmark of cerebral malaria (CM) is sequestration of Plasmodium falciparum-infected erythrocytes (IE) within the brain microvasculature. Binding of IE to endothelium reduces microvascular flow and, combined with an inflammatory response, perturbs endothelial barrier function, resulting in breakdown of the blood-brain barrier (BBB). Cytoadherence leads to activation of the endothelium and alters a range of cell processes affecting signaling pathways, receptor expression, coagulation, and disruption of BBB integrity. Here, we investigated whether CM-derived parasites elicit differential effects on human brain microvascular endothelial cells (HBMECs), as compared to uncomplicated malaria (UM)-derived parasites. Patient-derived IE from UM and CM clinical cases, as well as non-binding skeleton-binding protein 1 knockout parasites, were overlaid onto tumour necrosis factor (TNF)-activated HBMECs. Gene expression analysis of endothelial responses was performed using probe-based assays of a panel of genes involved in inflammation, apoptosis, endothelial barrier function, and prostacyclin synthesis pathway. We observed a significant effect on endothelial transcriptional responses in the presence of IE, yet there was no significant correlation between HBMEC responses and type of clinical syndrome (UM or CM). Furthermore, there was no correlation between HBMEC gene expression and both binding itself and level of IE binding to HBMECs, as we detected the same change in endothelial responses when employing both binding and non-binding parasites. Our results suggest that interaction of IE with endothelial cells in this co-culture model induces some endothelial responses that are independent of clinical origin and independent of the expression of the major variant antigen Plasmodium falciparum erythrocyte membrane protein 1 on the IE surface. IMPORTANCE: Cerebral malaria (CM) is the most prevalent and deadly complication of severe Plasmodium falciparum infection. A hallmark of this disease is sequestration of P. falciparum-infected erythrocytes (IE) in brain microvasculature that ultimately results in breakdown of the blood-brain barrier. Here, we compared the effect of P. falciparum parasites derived from uncomplicated malaria (UM) and CM cases on the relative gene expression of human brain microvascular endothelial cells (HBMECs) for a panel of genes. We observed a significant effect on the endothelial transcriptional response in the presence of IE, yet there is no significant correlation between HBMEC responses and the type of clinical syndrome (UM or CM). Furthermore, there was no correlation between HBMEC gene expression and both binding itself and the level of IE binding to HBMECs. Our results suggest that interaction of IE with endothelial cells induces endothelial responses that are independent of clinical origin and not entirely driven by surface Plasmodium falciparum erythrocyte membrane protein 1 expression.


Subject(s)
Blood-Brain Barrier , Brain , Endothelial Cells , Erythrocytes , Malaria, Cerebral , Malaria, Falciparum , Plasmodium falciparum , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Humans , Endothelial Cells/parasitology , Endothelial Cells/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism , Malaria, Cerebral/parasitology , Malaria, Cerebral/metabolism , Brain/parasitology , Brain/metabolism , Blood-Brain Barrier/parasitology , Blood-Brain Barrier/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism
16.
Antimicrob Agents Chemother ; 68(7): e0031124, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38874346

ABSTRACT

The emergence of clinically drug-resistant malaria parasites requires the urgent development of new drugs. Mosquitoes are vectors of multiple pathogens and have developed resistance mechanisms against them, which often involve antimicrobial peptides (AMPs). An-cecB is an AMP of the malaria-transmitting mosquito genus Anopheles, and we herein report its antimalarial activity against Plasmodium falciparum 3D7, the artemisinin-resistant strain 803, and the chloroquine-resistant strain Dd2 in vitro. We also demonstrate its anti-parasite activity in vivo, using the rodent malaria parasite Plasmodium berghei (ANKA). We show that An-cecB displays potent antimalarial activity and that its mechanism of action may occur through direct killing of the parasite or through interaction with infected red blood cell membranes. Unfortunately, An-cecB was found to be cytotoxic to mammalian cells and had poor antimalarial activity in vivo. However, its truncated peptide An-cecB-1 retained most of its antimalarial activity and avoided its cytotoxicity in vitro. An-cecB-1 also showed better antimalarial activity in vivo. Mosquito-derived AMPs may provide new ideas for the development of antimalarial drugs against drug-resistant parasites, and An-cecB has potential use as a template for antimalarial peptides.


Subject(s)
Anopheles , Antimalarials , Plasmodium berghei , Plasmodium falciparum , Animals , Antimalarials/pharmacology , Anopheles/drug effects , Anopheles/parasitology , Plasmodium falciparum/drug effects , Plasmodium berghei/drug effects , Mice , Cecropins/pharmacology , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Malaria/drug therapy , Malaria/parasitology , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Mosquito Vectors/drug effects , Mosquito Vectors/parasitology , Female , Insect Proteins/pharmacology , Drug Resistance/drug effects , Chloroquine/pharmacology , Parasitic Sensitivity Tests
17.
Infect Immun ; 92(7): e0001524, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38842304

ABSTRACT

Strain-transcending antibodies against virulence-associated subsets of P. falciparum-infected erythrocyte surface antigens could protect children from severe malaria. However, the evidence supporting the existence of such antibodies is incomplete and inconsistent. One subset of surface antigens associated with severe malaria, rosette-mediating Plasmodium falciparum Erythrocyte Membrane Protein one (PfEMP1) variants, cause infected erythrocytes to bind to uninfected erythrocytes to form clusters of cells (rosettes) that contribute to microvascular obstruction and pathology. Here, we tested plasma from 80 individuals living in malaria-endemic regions for IgG recognition of the surface of four P. falciparum rosetting strains using flow cytometry. Broadly reactive plasma samples were then used in antibody elution experiments in which intact IgG was eluted from the surface of infected erythrocytes and transferred to heterologous rosetting strains to look for strain-transcending antibodies. We found that seroprevalence (percentage of positive plasma samples) against allopatric rosetting strains was high in adults (63%-93%) but lower in children (13%-48%). Strain-transcending antibodies were present in nine out of eleven eluted antibody experiments, with six of these recognizing multiple heterologous rosetting parasite strains. One eluate had rosette-disrupting activity against heterologous strains, suggesting PfEMP1 as the likely target of the strain-transcending antibodies. Naturally acquired strain-transcending antibodies to rosetting P. falciparum strains in humans have not been directly demonstrated previously. Their existence suggests that such antibodies could play a role in clinical protection and raises the possibility that conserved epitopes recognized by strain-transcending antibodies could be targeted therapeutically by monoclonal antibodies or vaccines.


Subject(s)
Antibodies, Protozoan , Immunoglobulin G , Malaria, Falciparum , Plasmodium falciparum , Humans , Plasmodium falciparum/immunology , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Child , Adult , Immunoglobulin G/immunology , Immunoglobulin G/blood , Child, Preschool , Adolescent , Protozoan Proteins/immunology , Erythrocytes/parasitology , Erythrocytes/immunology , Antigens, Protozoan/immunology , Female , Male , Young Adult , Middle Aged , Seroepidemiologic Studies , Rosette Formation , Flow Cytometry
18.
PeerJ ; 12: e17406, 2024.
Article in English | MEDLINE | ID: mdl-38860213

ABSTRACT

Amphibians are experiencing declines globally, with emerging infectious diseases as one of the main causes. Haematological parameters present a useful method for determining the health status of animals and the effects of particular diseases, but the interpretation of differential cell counts relies on knowing the normal ranges for the species and factors that can affect these counts. However, there is very little data on either normal haematological parameters or guides for blood cell types for free-ranging frog species across the world. This study aims to 1) create a visual guide for three different Australian frog species: Litoria paraewingi, Limnodynastes dumerilii, and Crinia signifera, 2) determine the proportions of erythrocytes to leukocytes and 3) differential leukocytes within blood smears from these three species and 4) assess the association between parasites and differential counts. We collected blood samples from free-ranging frogs and analysed blood smears. We also looked for ectoparasites and tested for the fungal disease chytridiomycosis. Overall, we found that the differentials of erythrocytes to leukocytes were not affected by species, but the proportions of different leukocytes did vary across species. For example, while lymphocytes were the most common type of leukocyte across the three species, eosinophils were relatively common in Limnodynastes dumerilii but rarely present in the other two species. We noted chytridiomycosis infection as well as ectoparasites present in some individuals but found no effect of parasites on blood parameters. Our results add baseline haematological parameters for three Australian frog species and provide an example of how different frog species can vary in their differential blood cell counts. More information is needed on frog haematological data before these parameters can be used to determine the health status of wild or captive frogs.


Subject(s)
Anura , Animals , Anura/blood , Anura/parasitology , Anura/microbiology , Australia , Reference Values , Erythrocytes/parasitology , Blood Cell Count/veterinary , Hematologic Tests/veterinary , Species Specificity , Leukocyte Count , Male
19.
Front Immunol ; 15: 1350560, 2024.
Article in English | MEDLINE | ID: mdl-38863702

ABSTRACT

Background: Despite decades of effort, Plasmodium falciparum malaria remains a leading killer of children. The absence of a highly effective vaccine and the emergence of parasites resistant to both diagnosis as well as treatment hamper effective public health interventions. Methods and results: To discover new vaccine candidates, we used our whole proteome differential screening method and identified PfGBP130 as a parasite protein uniquely recognized by antibodies from children who had developed resistance to P. falciparum infection but not from those who remained susceptible. We formulated PfGBP130 as lipid encapsulated mRNA, DNA plasmid, and recombinant protein-based immunogens and evaluated the efficacy of murine polyclonal anti-PfGBP130 antisera to inhibit parasite growth in vitro. Immunization of mice with PfGBP130-A (aa 111-374), the region identified in our differential screen, formulated as a DNA plasmid or lipid encapsulated mRNA, but not as a recombinant protein, induced antibodies that inhibited RBC invasion in vitro. mRNA encoding the full ectodomain of PfGBP130 (aa 89-824) also generated parasite growth-inhibitory antibodies. Conclusion: We are currently advancing PfGBP130-A formulated as a lipid-encapsulated mRNA for efficacy evaluation in non-human primates.


Subject(s)
Antibodies, Protozoan , Erythrocytes , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Animals , Plasmodium falciparum/immunology , Antibodies, Protozoan/immunology , Mice , Erythrocytes/parasitology , Erythrocytes/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/parasitology , Humans , Malaria Vaccines/immunology , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Antigens, Protozoan/immunology , Immunization , Female
20.
Sci Rep ; 14(1): 14318, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38906910

ABSTRACT

Hemozoin is a natural biomarker formed during the hemoglobin metabolism of Plasmodium parasites, the causative agents of malaria. The rotating-crystal magneto-optical detection (RMOD) has been developed for its rapid and sensitive detection both in cell cultures and patient samples. In the current article we demonstrate that, besides quantifying the overall concentration of hemozoin produced by the parasites, RMOD can also track the size distribution of the hemozoin crystals. We establish the relations between the magneto-optical signal, the mean parasite age and the median crystal size throughout one erythrocytic cycle of Plasmodium falciparum parasites, where the latter two are determined by optical and scanning electron microscopy, respectively. The significant correlation between the magneto-optical signal and the stage distribution of the parasites indicates that the RMOD method can be utilized for species-specific malaria diagnosis and for the quick assessment of drug efficacy.


Subject(s)
Hemeproteins , Plasmodium falciparum , Hemeproteins/metabolism , Hemeproteins/chemistry , Plasmodium falciparum/growth & development , Humans , Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Malaria, Falciparum/diagnosis , Microscopy, Electron, Scanning/methods
SELECTION OF CITATIONS
SEARCH DETAIL