Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
PLoS One ; 17(1): e0263092, 2022.
Article in English | MEDLINE | ID: mdl-35077516

ABSTRACT

Knowledge about the relation of histopathological characteristics and mediators of physiological processes in the placenta malaria (PM) is poor, and that PM caused by Plasmodium vivax is almost null. The objective was to compare histopathological characteristics, cytokines and mediators of physiological processes in PM depending on the parasitic species, through a cross-sectional study in three groups: negative-PM, vivax-PM, falciparum-PM from Northwestern Colombia. The diagnosis of PM was made with thick blood smear, qPCR, and histopathology. Immuno-histochemical was made with EnVision system (Dako) and Zeiss Axio Imager M2 with light microscope. Cells in apoptosis were studied with the TUNEL technique. To measure the expression level of cytokines and mediators qRT-PCR was used. We included 179 placentas without PM and 87 with PM (53% P. vivax and 47% P. falciparum). At delivery, anemia was 25% in negative-PM, 60% in vivax-PM, and 44% in falciparum-PM group. The neonatal weight had an intense difference between groups with 3292±394g in negative-PM, 2,841±239 in vivax-PM, and 2,957±352 in falciparum-PM. The histopathological characteristics and CD+ cells in placenta with statistical differences (Dunn´s test) between negative-PM vs vivax-PM (P. falciparum was similar to P. vivax) were infarction, fibrinoid deposits, calcification, cells in apoptosis, immune infiltrates in decidua and intervillous space, CD4+, CD8+, CD14+, CD56+, CD68+. The expression levels of mediators in the placenta with statistical differences (Dunn´s test) between negative-PM vs vivax-PM (P. falciparum was similar to P. vivax) were Fas, FasL, HIF1α, Cox1, Cox2, VEGF, IL4, IL10, IFNγ, TNF, TGFß, FOXP3, and CTLA4. PM with P. falciparum and P. vivax, damages this organ and causes significant alteration of various physiological processes, which cause maternal anemia and a reduction in neonatal weight in degrees that are statistically and clinically significant. It is necessary that the search for plasmodial infection in pregnant and placenta goes from passive to active surveillance with adequate diagnostic capacity.


Subject(s)
Malaria, Falciparum/metabolism , Malaria, Vivax/metabolism , Placenta/metabolism , Plasmodium falciparum/metabolism , Plasmodium vivax/metabolism , Pregnancy Complications, Parasitic/metabolism , Adolescent , Adult , Colombia , Cytokines/metabolism , Female , Humans , Placenta/parasitology , Pregnancy , Pregnancy Complications, Parasitic/parasitology , Real-Time Polymerase Chain Reaction
2.
Nat Microbiol ; 6(8): 991-999, 2021 08.
Article in English | MEDLINE | ID: mdl-34294905

ABSTRACT

More than one-third of the world's population is exposed to Plasmodium vivax malaria, mainly in Asia1. P. vivax preferentially invades reticulocytes (immature red blood cells)2-4. Previous work has identified 11 parasite proteins involved in reticulocyte invasion, including erythrocyte binding protein 2 (ref. 5) and the reticulocyte-binding proteins (PvRBPs)6-10. PvRBP2b binds to the transferrin receptor CD71 (ref. 11), which is selectively expressed on immature reticulocytes12. Here, we identified CD98 heavy chain (CD98), a heteromeric amino acid transporter from the SLC3 family (also known as SLCA2), as a reticulocyte-specific receptor for the PvRBP2a parasite ligand using mass spectrometry, flow cytometry, biochemical and parasite invasion assays. We characterized the expression level of CD98 at the surface of immature reticulocytes (CD71+) and identified an interaction between CD98 and PvRBP2a expressed at the merozoite surface. Our results identify CD98 as an additional host membrane protein, besides CD71, that is directly associated with P. vivax reticulocyte tropism. These findings highlight the potential of using PvRBP2a as a vaccine target against P. vivax malaria.


Subject(s)
Erythrocytes/parasitology , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Malaria, Vivax/metabolism , Plasmodium vivax/metabolism , Antigens, CD , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Erythrocytes/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Host-Parasite Interactions , Humans , Malaria, Vivax/blood , Malaria, Vivax/genetics , Plasmodium vivax/genetics , Protein Binding , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Transferrin , Reticulocytes/metabolism , Reticulocytes/parasitology
3.
Commun Biol ; 3(1): 683, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33204009

ABSTRACT

Management of severe malaria remains a critical global challenge. In this study, using a multiplexed quantitative proteomics pipeline we systematically investigated the plasma proteome alterations in non-severe and severe malaria patients. We identified a few parasite proteins in severe malaria patients, which could be promising from a diagnostic perspective. Further, from host proteome analysis we observed substantial modulations in many crucial physiological pathways, including lipid metabolism, cytokine signaling, complement, and coagulation cascades in severe malaria. We propose that severe manifestations of malaria are possibly underpinned by modulations of the host physiology and defense machinery, which is evidently reflected in the plasma proteome alterations. Importantly, we identified multiple blood markers that can effectively define different complications of severe falciparum malaria, including cerebral syndromes and severe anemia. The ability of our identified blood markers to distinguish different severe complications of malaria may aid in developing new clinical tests for monitoring malaria severity.


Subject(s)
Malaria, Falciparum/diagnosis , Malaria, Falciparum/pathology , Proteomics/methods , Anemia/diagnosis , Anemia/pathology , Biomarkers/blood , Dengue/diagnosis , Dengue/metabolism , Dengue/pathology , Humans , Malaria, Falciparum/metabolism , Malaria, Vivax/blood , Malaria, Vivax/metabolism , Malaria, Vivax/pathology
4.
PLoS Negl Trop Dis ; 14(7): e0007656, 2020 07.
Article in English | MEDLINE | ID: mdl-32687542

ABSTRACT

Platelets drive endothelial cell activation in many diseases. However, if this occurs in Plasmodium vivax malaria is unclear. As platelets have been reported to be activated and to play a role in inflammatory response during malaria, we hypothesized that this would correlate with endothelial alterations during acute illness. We performed platelet flow cytometry of PAC-1 and P-selectin. We measured platelet markers (CXCL4, CD40L, P-selectin, Thrombopoietin, IL-11) and endothelial activation markers (ICAM-1, von Willebrand Factor and E-selectin) in plasma with a multiplex-based assay. The values of each mediator were used to generate heatmaps, K-means clustering and Principal Component analysis. In addition, we determined pair-wise Pearson's correlation coefficients to generate correlation networks. Platelet counts were reduced, and mean platelet volume increased in malaria patients. The activation of circulating platelets in flow cytometry did not differ between patients and controls. CD40L levels (Median [IQ]: 517 [406-651] vs. 1029 [732-1267] pg/mL, P = 0.0001) were significantly higher in patients, while P-selectin and CXCL4 showed a nonsignificant trend towards higher levels in patients. The network correlation approach demonstrated the correlation between markers of platelet and endothelial activation, and the heatmaps revealed a distinct pattern of activation in two subsets of P. vivax patients when compared to controls. Although absolute platelet activation was not strong in uncomplicated vivax malaria, markers of platelet activity and production were correlated with higher endothelial cell activation, especially in a specific subset of patients.


Subject(s)
Blood Platelets/cytology , Malaria, Vivax/blood , Adult , Blood Platelets/metabolism , CD40 Ligand/genetics , CD40 Ligand/metabolism , E-Selectin/genetics , E-Selectin/metabolism , Endothelial Cells/metabolism , Female , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-11/genetics , Interleukin-11/metabolism , Malaria, Vivax/genetics , Malaria, Vivax/metabolism , Male , P-Selectin/genetics , P-Selectin/metabolism , Platelet Activation , Platelet Count , Young Adult
5.
Cell Chem Biol ; 27(6): 719-727.e5, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32330444

ABSTRACT

Plasmodium vivax infects hepatocytes to form schizonts that cause blood infection, or dormant hypnozoites that can persist for months in the liver before leading to relapsing blood infections. The molecular processes that drive P. vivax schizont and hypnozoite survival remain largely unknown, but they likely involve a rich network of host-pathogen interactions, including those occurring at the host-parasite interface, the parasitophorous vacuole membrane (PVM). Using a recently developed P. vivax liver-stage model system we demonstrate that host aquaporin-3 (AQP3) localizes to the PVM of schizonts and hypnozoites within 5 days after invasion. This recruitment is also observed in P. vivax-infected reticulocytes. Chemical treatment with the AQP3 inhibitor auphen reduces P. vivax liver hypnozoite and schizont burden, and inhibits P. vivax asexual blood-stage growth. These findings reveal a role for AQP3 in P. vivax liver and blood stages and suggest that the protein may be targeted for therapeutic treatment.


Subject(s)
Aquaporin 3/metabolism , Liver/metabolism , Malaria, Vivax/metabolism , Plasmodium vivax/metabolism , Cells, Cultured , Humans , Liver/drug effects , Liver/parasitology , Malaria, Vivax/drug therapy , Malaria, Vivax/parasitology , Plasmodium vivax/isolation & purification
6.
Infect Immun ; 88(5)2020 04 20.
Article in English | MEDLINE | ID: mdl-32122938

ABSTRACT

Endothelial activation and microvascular dysfunction are key pathogenic processes in severe malaria. We evaluated the early role of these processes in experimentally induced Plasmodium falciparum and P. vivax infection. Participants were enrolled in induced blood-stage malaria clinical trials. Plasma osteoprotegerin, angiopoietin-2, and von Willebrand Factor (vWF) levels were measured as biomarkers of endothelial activation. Microvascular function was assessed using peripheral arterial tonometry and near-infrared spectroscopy, and the endothelial glycocalyx was assessed by sublingual videomicroscopy and measurement of biomarkers of degradation. Forty-five healthy, malaria-naive participants were recruited from 5 studies. Osteoprotegerin and vWF levels increased in participants following inoculation with P. vivax (n = 16) or P. falciparum (n = 15), with the angiopoietin-2 level also increasing in participants following inoculation with P. falciparum For both species, the most pronounced increase was seen in osteoprotegerin. This was particularly marked in participants inoculated with P. vivax, where the osteoprotegerin level correlated with the levels of parasitemia and the malaria clinical score. There were no changes in measures of endothelial glycocalyx or microvascular function. Plasma biomarkers of endothelial activation increased in early P. falciparum and P. vivax infection and preceded changes in the endothelial glycocalyx or microvascular function. The more pronounced increase in osteoprotegerin suggests that this biomarker may play a role in disease pathogenesis.


Subject(s)
Endothelial Cells/metabolism , Glycocalyx/metabolism , Malaria, Falciparum/metabolism , Malaria, Vivax/metabolism , Microvessels/metabolism , Plasmodium falciparum/pathogenicity , Plasmodium vivax/pathogenicity , Adolescent , Adult , Angiopoietin-2/metabolism , Biomarkers/metabolism , Female , Humans , Male , Middle Aged , Prospective Studies , Young Adult
7.
Malar J ; 19(1): 85, 2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32085712

ABSTRACT

BACKGROUND: Hypophosphatemia is common in severe infections including malaria. Previous studies suggested that serum phosphate concentrations correlate with temperature, but it is unclear whether the type of infection and other factors occurring during infection influence this association. Here relationships were investigated between serum phosphate levels, cause of fever, demographic, clinical and laboratory parameters. METHODS: Anonymized data were analysed from 633 adults with malaria or other febrile illness admitted to Northwick Park Hospital, London, UK. Univariable and multivariable generalized linear model analyses were performed to examine associations with serum phosphate levels. Interaction terms were included to investigate whether cause of fever (malaria vs other illness), malaria parasite species, or malaria severity influenced the association of other variables with phosphate. RESULTS: Hypophosphatemia was common in subjects with malaria (211/542 (39%)), and in other febrile illnesses (24/91 (26%)), however median phosphate levels did not differ significantly by diagnostic group, parasite species or severity of malaria. In all analyses, there were highly significant negative associations between serum phosphate and axillary temperature, and positive associations between serum phosphate and platelet count. There were no significant interactions between these variables and cause of fever, parasite species or severity of illness. Sodium and potassium concentrations were associated with serum phosphate in subjects with malaria and when data from all subjects was combined. CONCLUSION: Serum phosphate is consistently associated with temperature and platelet count in adults with diverse causes of fever. This may be a consequence of phosphate shifts from plasma into cells to support ATP generation for thermogenesis and platelet activation.


Subject(s)
Body Temperature , Malaria, Falciparum/metabolism , Malaria, Vivax/metabolism , Phosphates/blood , Adult , Diagnostic Tests, Routine , Female , Humans , London , Malaria, Falciparum/blood , Malaria, Vivax/blood , Male , Middle Aged , Young Adult
8.
PLoS Pathog ; 16(2): e1008258, 2020 02.
Article in English | MEDLINE | ID: mdl-32078643

ABSTRACT

The absence of the Duffy protein at the surface of erythrocytes was considered for decades to confer full protection against Plasmodium vivax as this blood group is the receptor for the key parasite ligand P. vivax Duffy binding protein (PvDBP). However, it is now clear that the parasite is able to break through this protection and induce clinical malaria in Duffy-negative people, although the underlying mechanisms are still not understood. Here, we briefly review the evidence of Duffy-negative infections by P. vivax and summarize the current hypothesis at the basis of this invasion process. We discuss those in the perspective of malaria-elimination challenges, notably in African countries.


Subject(s)
Antigens, Protozoan/metabolism , Duffy Blood-Group System/metabolism , Malaria, Vivax/metabolism , Plasmodium vivax , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Africa , Humans , Malaria, Vivax/prevention & control , Plasmodium vivax/metabolism , Plasmodium vivax/pathogenicity
9.
J Clin Invest ; 130(6): 2920-2927, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32045385

ABSTRACT

BACKGROUNDInterventions that interrupt Plasmodium vivax transmission or eliminate dormant P. vivax liver-stage parasites will be essential for malaria elimination. Development of these interventions has been hindered by the lack of P. vivax in vitro culture and could be accelerated by a safe and reproducible clinical model in malaria-naive individuals.METHODSHealthy, malaria-naive adults were enrolled in 2 studies to assess the safety, infectivity, and transmissibility of a new P. vivax isolate. Participants (Study 1, n = 2; Study 2, n = 24) were inoculated with P. vivax-infected red blood cells to initiate infection, and were treated with artemether-lumefantrine (Study 1) or chloroquine (Study 2). Primary endpoints were safety and infectivity of the new isolate. In Study 2, transmission to mosquitoes was also evaluated using mosquito feeding assays, and sporozoite viability was assessed using in vitro cultured hepatocytes.RESULTSParasitemia and gametocytemia developed in all participants and was cleared by antimalarial treatment. Adverse events were mostly mild or moderate and none were serious. Sixty-nine percent of participants (11/16) were infectious to Anopheles mosquitoes at peak gametocytemia. Mosquito infection rates reached 97% following membrane feeding with gametocyte-enriched blood, and sporozoites developed into liver-stage schizonts in culture.CONCLUSIONWe have demonstrated the safe, reproducible, and efficient transmission of P. vivax gametocytes from humans to mosquitoes, and have established an experimental model that will accelerate the development of interventions targeting multiple stages of the P. vivax life cycle.TRIAL REGISTRATIONACTRN12614000930684 and ACTRN12616000174482.FUNDING(Australian) National Health and Medical Research Council Program Grant 1132975 (Study 1). Bill and Melinda Gates Foundation (OPP1111147) (Study 2).


Subject(s)
Artemether, Lumefantrine Drug Combination/administration & dosage , Chloroquine/administration & dosage , Malaria, Vivax , Plasmodium vivax/metabolism , Adolescent , Adult , Animals , Anopheles , Female , Humans , Malaria, Vivax/drug therapy , Malaria, Vivax/metabolism , Malaria, Vivax/transmission , Male , Middle Aged , Models, Biological , Pilot Projects
10.
Immunol Rev ; 293(1): 163-189, 2020 01.
Article in English | MEDLINE | ID: mdl-31642531

ABSTRACT

Plasmodium vivax infection, the predominant cause of malaria in Asia and Latin America, affects ~14 million individuals annually, with considerable adverse effects on wellbeing and socioeconomic development. A clinical hallmark of Plasmodium infection, the paroxysm, is driven by pyrogenic cytokines produced during the immune response. Here, we review studies on the role of specific immune cell types, cognate innate immune receptors, and inflammatory cytokines on parasite control and disease symptoms. This review also summarizes studies on recurrent infections in individuals living in endemic regions as well as asymptomatic infections, a serious barrier to eliminating this disease. We propose potential mechanisms behind these repeated and subclinical infections, such as poor induction of immunological memory cells and inefficient T effector cells. We address the role of antibody-mediated resistance to P. vivax infection and discuss current progress in vaccine development. Finally, we review immunoregulatory mechanisms, such as inhibitory receptors, T regulatory cells, and the anti-inflammatory cytokine, IL-10, that antagonizes both innate and acquired immune responses, interfering with the development of protective immunity and parasite clearance. These studies provide new insights for the clinical management of symptomatic as well as asymptomatic individuals and the development of an efficacious vaccine for vivax malaria.


Subject(s)
Host-Parasite Interactions/immunology , Immunity , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Plasmodium vivax/physiology , Adaptive Immunity , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Cytokines/metabolism , Disease Susceptibility , Host-Parasite Interactions/genetics , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Malaria Vaccines/immunology , Malaria, Vivax/genetics , Malaria, Vivax/metabolism , Plasmodium vivax/growth & development , Toll-Like Receptors/metabolism
12.
Am J Hematol ; 94(9): 963-974, 2019 09.
Article in English | MEDLINE | ID: mdl-31148215

ABSTRACT

Malaria pathogenesis is caused by the replication of Plasmodium parasites within the red blood cells (RBCs) of the vertebrate host. This selective pressure has favored the evolution of protective polymorphisms in erythrocyte proteins, a subset of which serve as cognate receptors for parasite invasion ligands. Recently, the generation of RBCs from immortalized hematopoietic stem cells (HSCs) has offered a more tractable system for genetic manipulation and long-term in vitro culture, enabling elucidation of the functional determinants of host susceptibility in vitro. Here we report the generation of an immortalized erythroid progenitor cell line (EJ cells) from as few as 100 000 peripheral blood mononuclear cells. It offers a robust method for the creation of customized model systems from small volumes of peripheral blood. The EJ cell differentiation mirrored erythropoiesis of primary HSCs, yielding orthochromatic erythroblasts and enucleated RBCs after eight days (ejRBCs). The ejRBCs supported invasion by both P. vivax and P. falciparum. To demonstrate the genetic tractability of this system, we used CRISPR/Cas9 to disrupt the Duffy Antigen/Receptor for Chemokines (DARC) gene, which encodes the canonical receptor of P. vivax in humans. Invasion of P. vivax into this DARC-knockout cell line was strongly inhibited providing direct genetic evidence that P. vivax requires DARC for RBC invasion. Further, genetic complementation of DARC restored P. vivax invasion. Taken together, the peripheral blood immortalization method presented here offers the capacity to generate biologically representative model systems for studies of blood-stage malaria invasion from the peripheral blood of donors harboring unique genetic backgrounds, or rare polymorphisms.


Subject(s)
Erythroid Precursor Cells , Malaria, Falciparum , Malaria, Vivax , Models, Biological , Peripheral Blood Stem Cells , Plasmodium falciparum/metabolism , Plasmodium vivax/metabolism , Cell Line, Transformed , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/physiology , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Malaria, Vivax/metabolism , Malaria, Vivax/pathology , Peripheral Blood Stem Cells/metabolism , Peripheral Blood Stem Cells/parasitology , Peripheral Blood Stem Cells/pathology
13.
Proc Natl Acad Sci U S A ; 116(14): 7053-7061, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30872477

ABSTRACT

Unlike the case in Asia and Latin America, Plasmodium vivax infections are rare in sub-Saharan Africa due to the absence of the Duffy blood group antigen (Duffy antigen), the only known erythrocyte receptor for the P. vivax merozoite invasion ligand, Duffy binding protein 1 (DBP1). However, P. vivax infections have been documented in Duffy-negative individuals throughout Africa, suggesting that P. vivax may use ligands other than DBP1 to invade Duffy-negative erythrocytes through other receptors. To identify potential P. vivax ligands, we compared parasite gene expression in Saimiri and Aotus monkey erythrocytes infected with P. vivax Salvador I (Sal I). DBP1 binds Aotus but does not bind to Saimiri erythrocytes; thus, P. vivax Sal I must invade Saimiri erythrocytes independent of DBP1. Comparing RNA sequencing (RNAseq) data for late-stage infections in Saimiri and Aotus erythrocytes when invasion ligands are expressed, we identified genes that belong to tryptophan-rich antigen and merozoite surface protein 3 (MSP3) families that were more abundantly expressed in Saimiri infections compared with Aotus infections. These genes may encode potential ligands responsible for P. vivax infections of Duffy-negative Africans.


Subject(s)
Antigens, Protozoan/metabolism , Duffy Blood-Group System/metabolism , Erythrocytes/parasitology , Gene Expression Profiling , Malaria, Vivax/metabolism , Plasmodium vivax/metabolism , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, Protozoan/genetics , Duffy Blood-Group System/genetics , Erythrocytes/metabolism , Malaria, Vivax/genetics , Plasmodium vivax/genetics , Protozoan Proteins/genetics , Receptors, Cell Surface/genetics , Saimiri
14.
N Engl J Med ; 380(3): 215-228, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30650322

ABSTRACT

BACKGROUND: Treatment of Plasmodium vivax malaria requires the clearing of asexual parasites, but relapse can be prevented only if dormant hypnozoites are cleared from the liver (a treatment termed "radical cure"). Tafenoquine is a single-dose 8-aminoquinoline that has recently been registered for the radical cure of P. vivax. METHODS: This multicenter, double-blind, double-dummy, parallel group, randomized, placebo-controlled trial was conducted in Ethiopia, Peru, Brazil, Cambodia, Thailand, and the Philippines. We enrolled 522 patients with microscopically confirmed P. vivax infection (>100 to <100,000 parasites per microliter) and normal glucose-6-phosphate dehydrogenase (G6PD) activity (with normal activity defined as ≥70% of the median value determined at each trial site among 36 healthy male volunteers who were otherwise not involved in the trial). All patients received a 3-day course of chloroquine (total dose of 1500 mg). In addition, patients were assigned to receive a single 300-mg dose of tafenoquine on day 1 or 2 (260 patients), placebo (133 patients), or a 15-mg dose of primaquine once daily for 14 days (129 patients). The primary outcome was the Kaplan-Meier estimated percentage of patients who were free from recurrence at 6 months, defined as P. vivax clearance without recurrent parasitemia. RESULTS: In the intention-to-treat population, the percentage of patients who were free from recurrence at 6 months was 62.4% in the tafenoquine group (95% confidence interval [CI], 54.9 to 69.0), 27.7% in the placebo group (95% CI, 19.6 to 36.6), and 69.6% in the primaquine group (95% CI, 60.2 to 77.1). The hazard ratio for the risk of recurrence was 0.30 (95% CI, 0.22 to 0.40) with tafenoquine as compared with placebo (P<0.001) and 0.26 (95% CI, 0.18 to 0.39) with primaquine as compared with placebo (P<0.001). Tafenoquine was associated with asymptomatic declines in hemoglobin levels, which resolved without intervention. CONCLUSIONS: Single-dose tafenoquine resulted in a significantly lower risk of P. vivax recurrence than placebo in patients with phenotypically normal G6PD activity. (Funded by GlaxoSmithKline and Medicines for Malaria Venture; DETECTIVE ClinicalTrials.gov number, NCT01376167 .).


Subject(s)
Aminoquinolines/administration & dosage , Antimalarials/administration & dosage , Malaria, Vivax/drug therapy , Plasmodium vivax , Secondary Prevention/methods , Adolescent , Adult , Aminoquinolines/adverse effects , Antimalarials/adverse effects , Chloroquine/administration & dosage , Cytochrome P-450 CYP2D6/metabolism , Disease-Free Survival , Double-Blind Method , Drug Therapy, Combination , Female , Glucosephosphate Dehydrogenase/metabolism , Hemoglobins/analysis , Humans , Intention to Treat Analysis , Kaplan-Meier Estimate , Logistic Models , Malaria, Vivax/metabolism , Male , Parasitemia/drug therapy , Plasmodium vivax/isolation & purification , Primaquine/administration & dosage
15.
Malar J ; 18(1): 22, 2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30683097

ABSTRACT

BACKGROUND: Since malaria parasites highly depend on ribose 5-phosphate for DNA and RNA synthesis and on NADPH as a source of reducing equivalents, the pentose phosphate pathway (PPP) is considered an excellent anti-malarial drug target. In Plasmodium, a bifunctional enzyme named glucose 6-phosphate dehydrogenase 6-phosphogluconolactonase (GluPho) catalyzes the first two steps of the PPP. PfGluPho has been shown to be essential for the growth of blood stage Plasmodium falciparum parasites. METHODS: Plasmodium vivax glucose 6-phosphate dehydrogenase (PvG6PD) was cloned, recombinantly produced in Escherichia coli, purified, and characterized via enzyme kinetics and inhibitor studies. The effects of post-translational cysteine modifications were assessed via western blotting and enzyme activity assays. Genetically encoded probes were employed to study the effects of G6PD inhibitors on the cytosolic redox potential of Plasmodium. RESULTS: Here the recombinant production and characterization of PvG6PD, the C-terminal and NADPH-producing part of PvGluPho, is described. A comparison with PfG6PD (the NADPH-producing part of PfGluPho) indicates that the P. vivax enzyme has higher KM values for the substrate and cofactor. Like the P. falciparum enzyme, PvG6PD is hardly affected by S-glutathionylation and moderately by S-nitrosation. Since there are several naturally occurring variants of PfGluPho, the impact of these mutations on the kinetic properties of the enzyme was analysed. Notably, in contrast to many human G6PD variants, the mutations resulted in only minor changes in enzyme activity. Moreover, nanomolar IC50 values of several compounds were determined on P. vivax G6PD (including ellagic acid, flavellagic acid, and coruleoellagic acid), inhibitors that had been previously characterized on PfGluPho. ML304, a recently developed PfGluPho inhibitor, was verified to also be active on PvG6PD. Using genetically encoded probes, ML304 was confirmed to disturb the cytosolic glutathione-dependent redox potential of P. falciparum blood stage parasites. Finally, a new series of novel small molecules with the potential to inhibit the falciparum and vivax enzymes were synthesized, resulting in two compounds with nanomolar activity. CONCLUSION: The characterization of PvG6PD makes this enzyme accessible to further drug discovery activities. In contrast to naturally occurring G6PD variants in the human host that can alter the kinetic properties of the enzyme and thus the redox homeostasis of the cells, the naturally occurring PfGluPho variants studied here are unlikely to have a major impact on the parasites' redox homeostasis. Several classes of inhibitors have been successfully tested and are presently being followed up.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Glucosephosphate Dehydrogenase/genetics , Malaria, Vivax/genetics , Multienzyme Complexes/genetics , Protozoan Proteins/genetics , Carboxylic Ester Hydrolases/metabolism , Cloning, Molecular , Cytosol/metabolism , Escherichia coli/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/metabolism , Kinetics , Malaria, Vivax/enzymology , Malaria, Vivax/metabolism , Multienzyme Complexes/metabolism , Oxidation-Reduction , Protozoan Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
Science ; 359(6371): 48-55, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29302006

ABSTRACT

Plasmodium vivax shows a strict host tropism for reticulocytes. We identified transferrin receptor 1 (TfR1) as the receptor for P. vivax reticulocyte-binding protein 2b (PvRBP2b). We determined the structure of the N-terminal domain of PvRBP2b involved in red blood cell binding, elucidating the molecular basis for TfR1 recognition. We validated TfR1 as the biological target of PvRBP2b engagement by means of TfR1 expression knockdown analysis. TfR1 mutant cells deficient in PvRBP2b binding were refractory to invasion of P. vivax but not to invasion of P. falciparum Using Brazilian and Thai clinical isolates, we show that PvRBP2b monoclonal antibodies that inhibit reticulocyte binding also block P. vivax entry into reticulocytes. These data show that TfR1-PvRBP2b invasion pathway is critical for the recognition of reticulocytes during P. vivax invasion.


Subject(s)
Antigens, CD/metabolism , Malaria, Vivax/metabolism , Malaria, Vivax/parasitology , Membrane Proteins/chemistry , Plasmodium vivax/pathogenicity , Protozoan Proteins/chemistry , Receptors, Transferrin/metabolism , Reticulocytes/parasitology , Antigens, CD/genetics , Crystallography, X-Ray , Gene Knockdown Techniques , Host-Parasite Interactions , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Plasmodium vivax/metabolism , Protein Domains , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/ultrastructure , Receptors, Transferrin/genetics
19.
Proteomics Clin Appl ; 12(4): e1700046, 2018 07.
Article in English | MEDLINE | ID: mdl-28841253

ABSTRACT

Plasmodium vivax is the most geographically widespread species responsible for malaria in humans. Our study focused on identifying highly expressed parasite proteins using a shotgun proteomics approach. Parasites (P. vivax) are isolated from seven patient samples using saponin lysis. Protein extracts from these parasites are processed and subjected to LC-MS/MS analysis. An overall proteome coverage of 605 P. vivax proteins along with 1670 human host proteins are obtained upon combining the data from LC-MS/MS runs. While a major proportion of the P. vivax proteins are either hypothetical or involved in basic cellular activities, few proteins such as tryptophan-rich antigen (Pv-fam-a; PVX_090265), Pv-fam-d protein (PVX_101520), Plasmodium exported protein (PVX_003545), Pvstp1 (PVX_094303) and hypothetical protein (PVX_083555) are detected in more than 80% of the clinical isolates and found to be unique to P. vivax without orthologs in P. falciparum. Our proteomics study on individual parasite isolates reveals highly expressed P. vivax proteins, few of which may be good candidates for vivax malaria diagnosis due to their abundance and absence in P. falciparum. This study represents the first step towards the identification of biomarkers for P. vivax malaria. In future, their clinical diagnostic values must be explored and validated on large patient cohorts.


Subject(s)
Biomarkers/metabolism , Malaria, Vivax/metabolism , Plasmodium vivax/isolation & purification , Plasmodium vivax/metabolism , Proteome/metabolism , Proteomics/methods , Protozoan Proteins/metabolism , Humans , India/epidemiology , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Plasmodium vivax/pathogenicity
20.
Redox Biol ; 15: 192-206, 2018 05.
Article in English | MEDLINE | ID: mdl-29268202

ABSTRACT

Nitric oxide (NO) has dicotomic influence on modulating host-parasite interplay, synchronizing physiological orchestrations and diagnostic potential; instigated us to investigate the plausible association and genetic regulation among NO level, components of oxidative stress, iNOS polymorphisms and risk of malaria. Here, we experimentally elucidate that iNOS promoter polymorphisms are associated with risk of malaria; employing mutation specific genotyping, functional interplay using western blot and RT-PCR, quantitative estimation of NO, total antioxidant content (TAC) and reactive oxygen species (ROS). Genotyping revealed significantly associated risk of P. vivax (adjusted OR = 1.92 and 1.72) and P. falciparum (adjusted OR = 1.68 and 1.75) infection with SNP at iNOS-954G/C and iNOS-1173C/T positions, respectively; though vivax showed higher risk of infection. Intriguingly, mutation and infection specific differential upregulation of iNOS expression/NO level was observed and found to be significantly associated with mutant genotypes. Moreover, P. vivax showed pronounced iNOS protein (2.4 fold) and mRNA (2.5 fold) expression relative to healthy subjects. Furthermore, TAC and ROS were significantly decreased in infection; and differentially decreased in mutant genotypes. Our findings endorse polymorphic regulation of iNOS expression, altered oxidant-antioxidant components and evidences of risk association as the hallmark of malaria pathogenesis. iNOS/NO may serve as potential diagnostic marker in assessing clinical malaria.


Subject(s)
Host-Parasite Interactions/genetics , Malaria, Falciparum/genetics , Malaria, Vivax/genetics , Nitric Oxide Synthase Type II/genetics , Adult , Female , Genotype , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Malaria, Vivax/metabolism , Malaria, Vivax/parasitology , Malaria, Vivax/pathology , Male , Nitric Oxide/genetics , Nitric Oxide/metabolism , Oxidative Stress/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Plasmodium vivax/genetics , Plasmodium vivax/metabolism , Plasmodium vivax/pathogenicity , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...