Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.043
Filter
1.
Malar J ; 23(1): 267, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223522

ABSTRACT

BACKGROUND: The spread of antimalarial drug resistance parasites is a major obstacle in eliminating malaria in endemic areas. This increases the urgency for developing novel antimalarial drugs with improved profiles to eliminate both sensitive and resistant parasites in populations. The invention of the drug candidates needs a model for sensitive and resistant parasites on a laboratory scale. METHODS: Repeated Incomplete Treatment (RIcT) method was followed in raising the rodent malaria parasite, Plasmodium berghei, resistant to sulfadoxine. Plasmodium berghei were exposed to an adequate therapeutic dose of sulfadoxine without finishing the treatment to let the parasite recover. Cycles of drug treatment and parasite recovery were repeated until phenotypic resistance appeared. RESULTS: After undergoing 3-4 cycles, phenotypic resistance was not yet found in mice treated with sulfadoxine. Nevertheless, the molecular biology of dhps gene (the target of sulfadoxine) was analyzed at the end of the RIcT cycle. There was no mutations found in the gene target. Interestingly, the appearance of gametocytes at the end of every cycle of drug treatment and parasite recovery was observed. These gametocytes later on would no longer extend their life in the RBC stage, unless mosquitoes bite the infected host. This phenomenon is similar to the case in human malaria infections treated with sulfadoxine-pyrimethamine (SP). CONCLUSIONS: In this study, the antimalarial drug sulfadoxine induced gametocytogenesis in P. berghei, which could raise the risk factor for malaria transmission.


Subject(s)
Antimalarials , Plasmodium berghei , Sulfadoxine , Plasmodium berghei/drug effects , Antimalarials/pharmacology , Antimalarials/therapeutic use , Animals , Sulfadoxine/pharmacology , Sulfadoxine/therapeutic use , Mice , Drug Resistance/genetics , Gametogenesis/drug effects , Female , Malaria/drug therapy , Malaria/parasitology
2.
Sci Rep ; 14(1): 20344, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223194

ABSTRACT

Bovine lactoferrin (bLF) is a 77 kDa glycoprotein that is abundant in bovine breast milk and exerts various bioactive functions, including antibacterial and antiviral functions. Few studies have explored bLF activity against parasites. We found that bLF affects hemozoin synthesis by binding to heme, inhibiting heme iron polymerization necessary for Plasmodium berghei ANKA survival in infected erythrocytes, and also binds to hemozoin, causing it to disassemble. In a challenge test, bLF administration inhibited the growth of murine malaria parasites compared to untreated group growth. To determine whether the iron content of bLF affects the inhibition of malaria growth, we tested bLFs containing different amounts of iron (apo-bLF, native-bLF, and holo-bLF), but found no significant difference in their effects. This indicated that the active sites were located within the bLFs themselves. Further studies showed that the C-lobe domain of bLF can inhibit hemozoin formation and the growth of P. berghei ANKA. Evaluation of pepsin degradation products of the C-lobe identified a 47-amino-acid section, C-1, as the smallest effective region that could inhibit hemozoin formation. This study highlights bLF's potential as a novel therapeutic agent against malaria, underscoring the importance of its non-iron-dependent bioactive sites in combating parasite growth.


Subject(s)
Heme , Lactoferrin , Plasmodium berghei , Plasmodium berghei/drug effects , Plasmodium berghei/growth & development , Animals , Lactoferrin/pharmacology , Lactoferrin/metabolism , Cattle , Heme/metabolism , Mice , Hemeproteins/metabolism , Malaria/parasitology , Malaria/drug therapy , Protein Binding , Erythrocytes/parasitology , Erythrocytes/drug effects , Erythrocytes/metabolism , Iron/metabolism , Antimalarials/pharmacology
3.
Front Immunol ; 15: 1426316, 2024.
Article in English | MEDLINE | ID: mdl-39211036

ABSTRACT

γδ T cells facilitate the CD4+ T helper 1 (Th1) cell response against Plasmodium infection by activating conventional dendritic cells (cDCs), although the underlying mechanism remains elusive. Our study revealed that γδ T cells promote the complete maturation and production of interleukin-12 and CXCR3-ligands specifically in type 1 cDCs (cDC1), with minimal impact on cDC2 and monocyte derived DCs (Mo-DCs). During the initial infection phase, γδ T cell activation and temporal accumulation in the splenic white pulp, alongside cDC1, occur via CCR7-signaling. Furthermore, cDC1/γδ T cell interactions in the white pulp are amplified through CXCR3 signaling in γδ T cells, optimizing Th1 cell priming by cDC1. We also demonstrated how transitional Th1 cells arise in the white pulp before establishing their presence in the red pulp as fully differentiated Th1 cells. Additionally, we elucidate the reciprocal activation between γδ T cells and cDC1s. These findings suggest that Th1 cell priming is orchestrated by this reciprocal activation in the splenic white pulp during the early phase of blood-stage Plasmodium infection.


Subject(s)
Dendritic Cells , Lymphocyte Activation , Malaria , Th1 Cells , Th1 Cells/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Mice , Lymphocyte Activation/immunology , Malaria/immunology , Malaria/parasitology , Mice, Inbred C57BL , Receptors, CXCR3/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, CCR7/metabolism , Receptors, CCR7/immunology , Signal Transduction , Spleen/immunology , Cell Differentiation/immunology , Female
4.
Sci Rep ; 14(1): 20165, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39215071

ABSTRACT

Robust diagnostic tools and surveillance are crucial for malaria control and elimination efforts. Malaria caused by neglected Plasmodium parasites is often underestimated due to the lack of rapid diagnostic tools that can accurately detect these species. While nucleic-acid amplification technologies stand out as the most sensitive methods for detecting and confirming Plasmodium species, their implementation in resource-constrained settings poses significant challenges. Here, we present a Pan Plasmodium recombinase polymerase amplification lateral flow (RPA-LF) assay, capable of detecting all six human infecting Plasmodium species in low resource settings. The Pan Plasmodium RPA-LF assay successfully detected low density clinical infections with a preliminary limit of detection between 10-100 fg/µl for P. falciparum. When combined with crude nucleic acid extraction, the assay can serve as a point-of-need tool for molecular xenomonitoring. This utility was demonstrated by screening laboratory-reared Anopheles stephensi mosquitoes fed with Plasmodium-infected blood, as well as field samples of An. funestus s.l. and An. gambiae s.l. collected from central Africa. Overall, our proof-of-concept Pan Plasmodium diagnostic tool has the potential to be applied for clinical and xenomonitoring field surveillance, and after further evaluation, could become an essential tool to assist malaria control and elimination.


Subject(s)
Anopheles , Malaria , Mosquito Vectors , Nucleic Acid Amplification Techniques , Plasmodium , Humans , Animals , Anopheles/parasitology , Plasmodium/genetics , Plasmodium/isolation & purification , Nucleic Acid Amplification Techniques/methods , Malaria/diagnosis , Malaria/parasitology , Mosquito Vectors/parasitology , Recombinases/metabolism , Recombinases/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification
5.
Front Cell Infect Microbiol ; 14: 1438019, 2024.
Article in English | MEDLINE | ID: mdl-39149419

ABSTRACT

The malaria-causing parasites have to complete a complex infection cycle in the mosquito vector that also involves attack by the insect's innate immune system, especially at the early stages of midgut infection. However, Anopheles immunity to the late Plasmodium sporogonic stages, such as oocysts, has received little attention as they are considered to be concealed from immune factors due to their location under the midgut basal lamina and for harboring an elaborate cell wall comprising an external layer derived from the basal lamina that confers self-properties to an otherwise foreign structure. Here, we investigated whether Plasmodium berghei oocysts and sporozoites are susceptible to melanization-based immunity in Anopheles gambiae. Silencing of the negative regulator of melanization response, CLIPA14, increased melanization prevalence without significantly increasing the numbers of melanized oocysts, while co-silencing CLIPA14 with CLIPA2, a second negative regulator of melanization, resulted in a significant increase in melanized oocysts and melanization prevalence. Only late-stage oocysts were found to be melanized, suggesting that oocyst rupture was a prerequisite for melanization-based immune attack, presumably due to the loss of the immune-evasive features of their wall. We also found melanized sporozoites inside oocysts and in the hemocoel, suggesting that sporozoites at different maturation stages are susceptible to melanization. Silencing the melanization promoting factors TEP1 and CLIPA28 rescued oocyst melanization in CLIPA2/CLIPA14 co-silenced mosquitoes. Interestingly, silencing of CTL4, that protects early stage ookinetes from melanization, had no effect on oocysts and sporozoites, indicating differential regulation of immunity to early and late sporogonic stages. Similar to previous studies addressing ookinete stage melanization, the melanization of Plasmodium falciparum oocysts was significantly lower than that observed for P. berghei. In summary, our results provide conclusive evidence that late sporogonic malaria parasite stages are susceptible to melanization, and we reveal distinct regulatory mechanisms for ookinete and oocyst melanization.


Subject(s)
Anopheles , Melanins , Oocysts , Plasmodium berghei , Sporozoites , Animals , Anopheles/parasitology , Anopheles/immunology , Plasmodium berghei/immunology , Oocysts/metabolism , Melanins/metabolism , Sporozoites/immunology , Sporozoites/metabolism , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Insect Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/immunology , Malaria/immunology , Malaria/parasitology , Gene Silencing , Immunity, Innate , Female
6.
Nat Commun ; 15(1): 7105, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160174

ABSTRACT

Upon infecting its vertebrate host, the malaria parasite initially invades the liver where it undergoes massive replication, whilst remaining clinically silent. The coordination of host responses across the complex liver tissue during malaria infection remains unexplored. Here, we perform spatial transcriptomics in combination with single-nuclei RNA sequencing over multiple time points to delineate host-pathogen interactions across Plasmodium berghei-infected liver tissues. Our data reveals significant changes in spatial gene expression in the malaria-infected tissues. These include changes related to lipid metabolism in the proximity to sites of Plasmodium infection, distinct inflammation programs between lobular zones, and regions with enrichment of different inflammatory cells, which we term 'inflammatory hotspots'. We also observe significant upregulation of genes involved in inflammation in the control liver tissues of mice injected with mosquito salivary gland components. However, this response is considerably delayed compared to that observed in P. berghei-infected mice. Our study establishes a benchmark for investigating transcriptome changes during host-parasite interactions in tissues, it provides informative insights regarding in vivo study design linked to infection and offers a useful tool for the discovery and validation of de novo intervention strategies aimed at malaria liver stage infection.


Subject(s)
Liver , Malaria , Plasmodium berghei , Animals , Liver/parasitology , Liver/metabolism , Plasmodium berghei/physiology , Malaria/parasitology , Mice , Host-Pathogen Interactions , Transcriptome , Host-Parasite Interactions , Single-Cell Analysis , Mice, Inbred C57BL , Female , Inflammation , Gene Expression Profiling , Lipid Metabolism
7.
Nat Commun ; 15(1): 7487, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39209843

ABSTRACT

CelTOS is a malaria vaccine antigen that is conserved in Plasmodium and other apicomplexan parasites and plays a role in cell-traversal. The structural basis and mechanisms of CelTOS-induced protective immunity to parasites are unknown. Here, CelTOS-specific monoclonal antibodies (mAbs) 7g7 and 4h12 demonstrated multistage activity, protecting against liver infection and preventing parasite transmission to mosquitoes. Both mAbs demonstrated cross-species activity with sterile protection against in vivo challenge with transgenic parasites containing either P. falciparum or P. vivax CelTOS, and with transmission reducing activity against P. falciparum. The mAbs prevented CelTOS-mediated pore formation providing insight into the protective mechanisms. X-ray crystallography and mutant-library epitope mapping revealed two distinct broadly conserved neutralizing epitopes. 7g7 bound to a parallel dimer of CelTOS, while 4h12 bound to a novel antiparallel dimer architecture. These findings inform the design of antibody therapies and vaccines and raise the prospect of a single intervention to simultaneously combat P. falciparum and P. vivax malaria.


Subject(s)
Antibodies, Monoclonal , Antibodies, Protozoan , Malaria Vaccines , Plasmodium falciparum , Plasmodium vivax , Antibodies, Monoclonal/immunology , Animals , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Malaria Vaccines/immunology , Antibodies, Protozoan/immunology , Mice , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/parasitology , Crystallography, X-Ray , Epitopes/immunology , Malaria, Vivax/prevention & control , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Antigens, Protozoan/immunology , Humans , Female , Epitope Mapping , Malaria/immunology , Malaria/prevention & control , Malaria/parasitology , Mice, Inbred BALB C , Protozoan Proteins/immunology , Protozoan Proteins/chemistry
8.
Emerg Infect Dis ; 30(9): 1884-1894, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39174028

ABSTRACT

Malaria is increasingly diagnosed in urban centers across the Amazon Basin. In this study, we combined repeated prevalence surveys over a 4-year period of a household-based random sample of 2,774 persons with parasite genotyping to investigate the epidemiology of malaria in Mâncio Lima, the main urban transmission hotspot in Amazonian Brazil. We found that most malarial infections were asymptomatic and undetected by point-of-care microscopy. Our findings indicate that as malaria transmission decreases, the detection threshold of microscopy rises, resulting in more missed infections despite similar parasite densities estimated by molecular methods. We identified genetically highly diverse populations of Plasmodium vivax and P. falciparum in the region; occasional shared lineages between urban and rural residents suggest cross-boundary propagation. The prevalence of low-density and asymptomatic infections poses a significant challenge for routine surveillance and the effectiveness of malaria control and elimination strategies in urbanized areas with readily accessible laboratory facilities.


Subject(s)
Microscopy , Brazil/epidemiology , Humans , Prevalence , Microscopy/methods , Female , Male , Adult , Adolescent , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Child , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Malaria/epidemiology , Malaria/transmission , Malaria/prevention & control , Malaria/parasitology , Plasmodium vivax/genetics , Urban Population , Child, Preschool , Plasmodium falciparum/genetics , Middle Aged , Young Adult , Infant , History, 21st Century
9.
Infect Genet Evol ; 123: 105650, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089500

ABSTRACT

Despite the critical role of the Anopheles innate immune system in defending against Plasmodium infection, there is still limited information about the key immune mechanisms in Anopheles. This review assesses recent findings on the expression characteristics of immune-related genes in Anopheles following exposure to Plasmodium. A literature review, unrestricted by publication date, was conducted to evaluate immune-related gene expression in different organs of Anopheles after Plasmodium infection. Mosquito immune responses in the midgut are essential for reducing parasite populations. Additionally, innate immune responses in the salivary glands and hemocytes circulating in the hemocoel play key roles in defense against the parasite. Transcriptomic analysis of the mosquito's innate immune response to Plasmodium infection provides valuable insights into key immune mechanisms in mosquito defense. A deeper understanding of immune mechanisms in different organs of Anopheles following Plasmodium infection will aid in discovering critical targets for designing novel control strategies.


Subject(s)
Anopheles , Immunity, Innate , Malaria , Plasmodium , Animals , Anopheles/parasitology , Anopheles/genetics , Anopheles/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium/immunology , Plasmodium/genetics , Gene Expression Profiling , Mosquito Vectors/parasitology , Mosquito Vectors/genetics , Mosquito Vectors/immunology , Host-Parasite Interactions/immunology , Transcriptome
10.
Int Immunopharmacol ; 140: 112843, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39098224

ABSTRACT

Neutrophils and macrophages confine pathogens by entrapping them in extracellular traps (ETs) through activating TLR9 function. However, plasmodial parasites secreted TatD-like DNases (TatD) to counteract ETs-mediated immune clearance. We found that TLR9 mutant mice increased susceptibility to rodent malaria, suggesting TLR9 is a key protein for host defense. We found that the proportion of neutrophils and macrophages in response to plasmodial parasite infection in the TLR9 mutant mice was significantly reduced compared to that of the WT mice. Importantly, PbTatD can directly bind to the surface TLR9 (sTLR9) on macrophages, which blocking the phosphorylation of mitogen-activated protein kinase and nuclear factor-κB, negatively regulated the signaling of ETs formation by both macrophages and neutrophils. Such, P. berghei TatD is a parasite virulence factor that can inhibit the proliferation of macrophages and neutrophils through directly binding to TLR9 receptors on the cell surface, thereby blocking the activation of the downstream MyD88-NF-kB pathways.


Subject(s)
Deoxyribonucleases , Immunity, Innate , Macrophages , Malaria , Mice, Inbred C57BL , NF-kappa B , Neutrophils , Plasmodium berghei , Signal Transduction , Toll-Like Receptor 9 , Toll-Like Receptor 9/metabolism , Animals , NF-kappa B/metabolism , Plasmodium berghei/immunology , Neutrophils/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Malaria/immunology , Malaria/parasitology , Deoxyribonucleases/metabolism , Extracellular Traps/immunology , Extracellular Traps/metabolism , Mice, Knockout , Protozoan Proteins/metabolism , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Humans
11.
Cell Rep ; 43(8): 114600, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39126653

ABSTRACT

Malaria is initiated as Plasmodium sporozoites are injected into the dermis when an infected mosquito probes on a vertebrate host for a blood meal. Factors in the mosquito saliva, such as AgTRIO, can alter the ability of Anopheles gambiae to transmit Plasmodium. We therefore used CRISPR-Cas9-mediated genome editing to generate AgTRIO knockout (KO) A. gambiae and examined the ability of these mosquitoes to probe on a vertebrate host. AgTRIO KO mosquitoes showed a diminished host probing capacity and required repetitive probing to locate a blood resource to complete a blood meal. This increased probing resulted in enhanced Plasmodium transmission to the vertebrate host. Our data demonstrate the importance of the A. gambiae saliva protein AgTRIO in probing and its influence on the ability of mosquitoes to transmit malaria.


Subject(s)
Anopheles , Animals , Anopheles/parasitology , Anopheles/genetics , Malaria/transmission , Malaria/parasitology , Insect Proteins/genetics , Insect Proteins/metabolism , Mice , CRISPR-Cas Systems/genetics , Female , Mosquito Vectors/parasitology , Mosquito Vectors/genetics
12.
Parasitol Res ; 123(8): 308, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39172181

ABSTRACT

Malaria has a historical presence in the Dakshina Kannada (D.K.) and Udupi districts of Karnataka, India. To understand the potential involvement of anopheline fauna in malaria transmission, we conducted an exploratory entomological survey. The study is crucial given the decreasing malaria incidence in these districts in recent years. From September 2022 to August 2023, we collected indoor resting mosquitoes using a manual aspirator at 27 randomly chosen sites within three distinct resting habitats (human dwellings, cattle sheds, and construction sites) in the urban areas of Udupi and Dakshina Kannada districts. Mosquitoes were morphologically identified, and anopheline specimens were tested for the presence of malarial parasite by polymerase chain reaction (PCR) analysis. We collected a total of 1810 mosquitoes, comprising 21 species distributed across five genera. Culex emerged as the predominant genus, constituting 84.4% of the collected specimens, while Anopheles accounted for 5.4%. Among the observed species, Culex quinquefasciatus was predominant, comprising 77.9% of the mosquito specimens collected in this study. Two malaria vectors, An. stephensi and An. subpictus complex, constituted 16.3% and 1.0% of the total anophelines collected, respectively. None of the 96 female anophelines was tested positive for Plasmodium infection. Our findings suggest that Anopheles mosquitoes prefer resting in cattle sheds over human dwellings. While our study identified two malaria vectors, they were present at low densities. To gain a more comprehensive understanding of the dynamics of these vector mosquitoes, it is essential to conduct long-term surveillance to monitor their prevalence and role in malaria transmission.


Subject(s)
Anopheles , Ecosystem , Malaria , Mosquito Vectors , Animals , India/epidemiology , Anopheles/parasitology , Anopheles/physiology , Anopheles/classification , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Malaria/transmission , Malaria/epidemiology , Malaria/parasitology , Humans , Prevalence , Plasmodium/isolation & purification , Plasmodium/classification , Plasmodium/physiology , Cattle , Female , Culex/parasitology , Culex/physiology
13.
Nat Commun ; 15(1): 7206, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174515

ABSTRACT

Apical membrane antigen-1 (AMA1) is a conserved malarial vaccine candidate essential for the formation of tight junctions with the rhoptry neck protein (RON) complex, enabling Plasmodium parasites to invade human erythrocytes, hepatocytes, and mosquito salivary glands. Despite its critical role, extensive surface polymorphisms in AMA1 have led to strain-specific protection, limiting the success of AMA1-based interventions beyond initial clinical trials. Here, we identify an i-body, a humanised single-domain antibody-like molecule that recognises a conserved pan-species conformational epitope in AMA1 with low nanomolar affinity and inhibits the binding of the RON2 ligand to AMA1. Structural characterisation indicates that the WD34 i-body epitope spans the centre of the conserved hydrophobic cleft in AMA1, where interacting residues are highly conserved among all Plasmodium species. Furthermore, we show that WD34 inhibits merozoite invasion of erythrocytes by multiple Plasmodium species and hepatocyte invasion by P. falciparum sporozoites. Despite a short half-life in mouse serum, we demonstrate that WD34 transiently suppressed P. berghei infections in female BALB/c mice. Our work describes the first pan-species AMA1 biologic with inhibitory activity against multiple life-cycle stages of Plasmodium. With improved pharmacokinetic characteristics, WD34 could be a potential immunotherapy against multiple species of Plasmodium.


Subject(s)
Antigens, Protozoan , Erythrocytes , Liver , Membrane Proteins , Mice, Inbred BALB C , Protozoan Proteins , Animals , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Female , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Humans , Erythrocytes/parasitology , Erythrocytes/immunology , Liver/parasitology , Liver/immunology , Liver/metabolism , Malaria Vaccines/immunology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Cross Reactions/immunology , Plasmodium falciparum/immunology , Plasmodium berghei/immunology , Epitopes/immunology , Hepatocytes/parasitology , Hepatocytes/immunology , Hepatocytes/metabolism , Plasmodium/immunology , Merozoites/immunology , Merozoites/metabolism
14.
PLoS Negl Trop Dis ; 18(8): e0012424, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39150978

ABSTRACT

The risk of severe malaria from the zoonotic parasite Plasmodium knowlesi approximates that from P. falciparum. In severe falciparum malaria, neutrophil activation contributes to inflammatory pathogenesis, including acute lung injury (ALI). The role of neutrophil activation in the pathogenesis of severe knowlesi malaria has not been examined. We evaluated 213 patients with P. knowlesi mono-infection (138 non-severe, 75 severe) and 49 Plasmodium-negative controls from Malaysia. Markers of neutrophil activation (soluble neutrophil elastase [NE], citrullinated histone [CitH3] and circulating neutrophil extracellular traps [NETs]) were quantified in peripheral blood by microscopy and immunoassays. Findings were correlated with malaria severity, ALI clinical criteria, biomarkers of parasite biomass, haemolysis, and endothelial activation. Neutrophil activation increased with disease severity, with median levels higher in severe than non-severe malaria and controls for NE (380[IQR:210-930]ng/mL, 236[139-448]ng/mL, 218[134-307]ng/mL, respectively) and CitH3 (8.72[IQR:3.0-23.1]ng/mL, 4.29[1.46-9.49]ng/mL, 1.53[0.6-2.59]ng/mL, respectively)[all p<0.01]. NETs were higher in severe malaria compared to controls (126/µL[IQR:49-323] vs 51[20-75]/µL, p<0.001). In non-severe malaria, neutrophil activation fell significantly upon discharge from hospital (p<0.03). In severe disease, NETs, NE, and CitH3 were correlated with parasitaemia, cell-free haemoglobin and angiopoietin-2 (all Pearson's r>0.24, p<0.05). Plasma NE and angiopoietin-2 were higher in knowlesi patients with ALI than those without (p<0.008); neutrophilia was associated with an increased risk of ALI (aOR 3.27, p<0.01). In conclusion, neutrophil activation is increased in ALI and in proportion to disease severity in knowlesi malaria, is associated with endothelial activation, and may contribute to disease pathogenesis. Trials of adjunctive therapies to regulate neutrophil activation are warranted in severe knowlesi malaria.


Subject(s)
Acute Lung Injury , Extracellular Traps , Malaria , Neutrophil Activation , Neutrophils , Plasmodium knowlesi , Severity of Illness Index , Humans , Male , Female , Malaria/immunology , Malaria/blood , Malaria/parasitology , Adult , Acute Lung Injury/immunology , Acute Lung Injury/parasitology , Acute Lung Injury/pathology , Middle Aged , Neutrophils/immunology , Extracellular Traps/immunology , Malaysia , Biomarkers/blood , Young Adult , Leukocyte Elastase/blood , Histones/blood , Adolescent
15.
Curr Top Membr ; 93: 27-49, 2024.
Article in English | MEDLINE | ID: mdl-39181577

ABSTRACT

Malaria remains a major global threat, representing a severe public health problem worldwide. Annually, it is responsible for a high rate of morbidity and mortality in many tropical developing countries where the disease is endemic. The causative agent of malaria, Plasmodium spp., exhibits a complex life cycle, alternating between an invertebrate vector, which transmits the disease, and the vertebrate host. The disease pathology observed in the vertebrate host is attributed to the asexual development of Plasmodium spp. inside the erythrocyte. Once inside the red blood cell, malaria parasites cause extensive changes in the host cell, increasing membrane rigidity and altering its normal discoid shape. Additionally, during their intraerythrocytic development, malaria parasites incorporate and degrade up to 70 % of host cell hemoglobin. This mechanism is essential for parasite development and represents an important drug target. Blocking the steps related to hemoglobin endocytosis or degradation impairs parasite development and can lead to its death. The ultrastructural analysis of hemoglobin endocytosis on Plasmodium spp. has been broadly explored along the years. However, it is only recently that the proteins involved in this process have started to emerge. Here, we will review the most important features related to hemoglobin endocytosis and catabolism on malaria parasites. A special focus will be given to the recent analysis obtained through 3D visualization approaches and to the molecules involved in these mechanisms.


Subject(s)
Endocytosis , Malaria , Plasmodium , Animals , Humans , Malaria/parasitology , Malaria/metabolism , Plasmodium/metabolism , Plasmodium/physiology , Erythrocytes/parasitology , Erythrocytes/metabolism , Cell Membrane/metabolism , Hemoglobins/metabolism
16.
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
17.
Acta Trop ; 258: 107350, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39134111

ABSTRACT

Malaria and babesiosis are global health threats affecting humans, wildlife, and domestic animals, particularly in Africa, the Americas, and Europe. Malaria can lead to severe outcomes, while babesiosis usually resembles a mild illness but can be severe and fatal in individuals with weakened immune systems. Swift, accurate detection of these parasites is crucial for treatment and control. We evaluated a real-time PCR assay for diagnosing five Plasmodium and three Babesia species from blood samples, assessing its sensitivity, specificity, and analytical performance by analyzing 46 malaria-positive and 32 Babesia spp-positive samples diagnosed through microscopy. The limit of detection for Plasmodium species ranged from 30 to 0.0003 copies/µL. For mixed infections, it was 0.3 copies/µL for P. falciparum/P. vivax and 3 copies/µL for P. malariae/P. knowlesi. Babesia species had a detection limit of 0.2 copies/µL. No cross-reactivity was observed among 64 DNA samples from various microorganisms. The assay showed good sensitivity, detecting Plasmodium and Babesia species with 100 % accuracy overall, except for P. falciparum (97.7 %) and B. microti (12.5 %). The low sensitivity of detecting B. microti was attributed to limitations in microscopy for species identification. This technique heavily relies on the proficiency of the examiner, as species within the genus cannot be distinguished under a microscope. Additionally, Babesia can be confused with the early trophozoite stage (ring forms) of Plasmodium parasites. The findings support multiplex qPCR's diagnostic superiority over the gold standard, despite higher costs. It offers enhanced sensitivity, specificity, and detects mixed infections, crucial for effective monitoring and diagnosis of malaria and babesiosis in endemic regions with significant public health challenges.


Subject(s)
Babesia , Babesiosis , DNA, Protozoan , Malaria , Plasmodium , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Real-Time Polymerase Chain Reaction/methods , Babesia/genetics , Babesia/isolation & purification , Babesia/classification , Plasmodium/isolation & purification , Plasmodium/genetics , Plasmodium/classification , Humans , Malaria/diagnosis , Malaria/parasitology , Babesiosis/diagnosis , Babesiosis/parasitology , Babesiosis/blood , DNA, Protozoan/genetics , DNA, Protozoan/blood
18.
Front Cell Infect Microbiol ; 14: 1405198, 2024.
Article in English | MEDLINE | ID: mdl-39108983

ABSTRACT

Introduction: Plasmodium malariae is the most common non-falciparum species in sub-Saharan Africa. Despite this, data on its genetic diversity is scarce. Therefore, we aimed to establish a P. malariae genotyping approach based on size polymorphic regions that can be easily applied in molecular epidemiological studies. Methods: Four potential genotyping markers, Pm02, Pm09, P. malariae thrombospondin-related anonymous protein (pmtrap), and P. malariae merozoite surface protein fragment 2 (pmmsp1 F2) were amplified via nested PCR and analysed using automated capillary gel electrophoresis. Results: We observed the highest allelic diversity for pmtrap (MOI = 1.61) and pmmsp1 F2 (He = 0.81). Further applying the two markers pmtrap and pmmsp1 F2 on a different sample set of 21 P. malariae positive individuals followed up over one week, we saw a high consistency in their performance. The results show a large complexity and high dynamics of P. malariae infections in the asymptomatic Gabonese study population. Discussion: We successfully implemented a new genotyping panel for P. malariae consisting of only two markers: pmtrap and pmmsp1 F2. It can be easily applied in other settings to investigate the genotype diversity of P. malariae populations, providing further important data on the molecular epidemiology of this parasite species.


Subject(s)
Genetic Variation , Genotype , Malaria , Molecular Epidemiology , Plasmodium malariae , Protozoan Proteins , Plasmodium malariae/genetics , Plasmodium malariae/isolation & purification , Humans , Malaria/epidemiology , Malaria/parasitology , Molecular Epidemiology/methods , Africa South of the Sahara/epidemiology , Protozoan Proteins/genetics , Genotyping Techniques/methods , Polymerase Chain Reaction/methods , DNA, Protozoan/genetics , Alleles , Gabon/epidemiology , Genetic Markers
19.
Sci Rep ; 14(1): 18162, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39107355

ABSTRACT

Malaria has complex interactions with host physiology, including alterations in cortisol levels. Cortisol, a key hormone in the stress response, is known to be dysregulated in various infectious diseases. This systematic review and meta-analysis aimed to elucidate the relationship between Plasmodium infection and cortisol levels, shedding light on the intricate interplay between the parasite and the host's endocrine system. The methodological protocol for assessing cortisol levels in malaria patients was registered in PROSPERO (CRD42024496578), a widely recognized international prospective register of systematic reviews. This registration ensures transparency and minimizes the risk of bias in our research. A comprehensive search strategy was employed across major databases, including Embase, PubMed, Scopus, and Medline, to include studies that reported cortisol levels in infected patients. The qualitative synthesis was undertaken to synthesize the difference in cortisol levels between malaria-infected and uninfected individuals. The meta-analysis employed the random effects model in the quantitative synthesis to calculate the effect estimate. The review included a total of 20 studies, with a substantial number conducted in Africa, followed by Asia and South America. Most included studies (13/20, 65%) reported higher cortisol levels in infected patients than in uninfected patients. The meta-analysis confirmed significantly higher cortisol levels in infected patients compared to uninfected individuals (P < 0.0001, standardized mean difference (SMD): 1.354, 95% confidence interval: 0.913 to 1.795, I2: 88.3%, across 15 studies). Notably, the method for cortisol measurement and the type of blood sample used (serum or plasma) were significant moderators in the analysis, indicating that these factors may influence the observed relationship between Plasmodium infection and cortisol levels. The systematic review and meta-analysis confirmed that Plasmodium infection is associated with increased cortisol levels, highlighting the intricate relationship between the disease and the host stress response. These findings underscore the potential of cortisol as a supplementary biomarker for understanding the pathophysiological impact of malaria. By providing insights into the stress-related mechanisms of malaria, this comprehensive understanding can inform future research and potentially enhance disease management and treatment strategies, particularly in regions heavily burdened by malaria.


Subject(s)
Hydrocortisone , Malaria , Hydrocortisone/blood , Hydrocortisone/metabolism , Humans , Malaria/blood , Malaria/metabolism , Malaria/parasitology , Plasmodium
20.
PLoS Comput Biol ; 20(8): e1012327, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102445

ABSTRACT

Plasmodium parasites cause Malaria disease, which remains a significant threat to global health, affecting 200 million people and causing 400,000 deaths yearly. Plasmodium falciparum and Plasmodium vivax remain the two main malaria species affecting humans. Identifying the malaria disease in blood smears requires years of expertise, even for highly trained specialists. Literature studies have been coping with the automatic identification and classification of malaria. However, several points must be addressed and investigated so these automatic methods can be used clinically in a Computer-aided Diagnosis (CAD) scenario. In this work, we assess the transfer learning approach by using well-known pre-trained deep learning architectures. We considered a database with 6222 Region of Interest (ROI), of which 6002 are from the Broad Bioimage Benchmark Collection (BBBC), and 220 were acquired locally by us at Fundação Oswaldo Cruz (FIOCRUZ) in Porto Velho Velho, Rondônia-Brazil, which is part of the legal Amazon. We exhaustively cross-validated the dataset using 100 distinct partitions with 80% train and 20% test for each considering circular ROIs (rough segmentation). Our experimental results show that DenseNet201 has a potential to identify Plasmodium parasites in ROIs (infected or uninfected) of microscopic images, achieving 99.41% AUC with a fast processing time. We further validated our results, showing that DenseNet201 was significantly better (99% confidence interval) than the other networks considered in the experiment. Our results support claiming that transfer learning with texture features potentially differentiates subjects with malaria, spotting those with Plasmodium even in Leukocytes images, which is a challenge. In Future work, we intend scale our approach by adding more data and developing a friendly user interface for CAD use. We aim at aiding the worldwide population and our local natives living nearby the legal Amazon's rivers.


Subject(s)
Microscopy , Humans , Microscopy/methods , Plasmodium falciparum/pathogenicity , Plasmodium vivax , Computational Biology/methods , Malaria/parasitology , Plasmodium , Deep Learning , Databases, Factual , Image Processing, Computer-Assisted/methods , Malaria, Falciparum/parasitology , Diagnosis, Computer-Assisted/methods
SELECTION OF CITATIONS
SEARCH DETAIL