Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.971
Filter
1.
PLoS One ; 19(7): e0306664, 2024.
Article in English | MEDLINE | ID: mdl-38968270

ABSTRACT

BACKGROUNDS: Malaria, a preventive and treatable disease, is still responsible for annual deaths reported in most tropical regions, principally in sub-Saharan Africa. Subunit recombinant transmission-blocking vaccines (TBVs) have been proposed as promising vaccines to succeed in malaria elimination and eradication. Here, a provisional study was designed to assess the immunogenicity and functional activity of alanyl aminopeptidase N (APN1) of Anopheles stephensi, as a TBV candidate, administered with MPL, CpG, and QS21 adjuvants in the murine model. METHODOLOGY/PRINCIPAL FINDINGS: The mouse groups were immunized with recombinant APN1 (rAPN1) alone or formulated with CpG, MPL, QS-21, or a combination of adjuvants (CMQ), and the elicited immune responses were evaluated after the third immunization. The standard membrane feeding assay (SMFA) measured the functional activity of antibodies against bacterial-expressed APN1 protein in adjuvanted vaccine groups on transmission of P. falciparum (NF54) to An. stephensi mosquitoes. Evaluation of mice vaccinated with rAPN1 formulated with distinct adjuvants manifested a significant increase in the high-avidity level of anti-APN1 IgG and IgG subclasses; however, rAPN1 induced the highest level of high-avidity anti-APN1 IgG1, IgG2a, and IgG2b antibodies in the immunized vaccine group 5 (APN1/CMQ). In addition, vaccine group 5 (receiving APN1/CMQ), had still the highest level of anti-APN1 IgG antibodies relative to other immunized groups after six months, on day 180. The SMFA data indicates a trend towards higher transmission-reducing activity in groups 2 and 5, which received the antigen formulated with CpG or a combination of three adjuvants. CONCLUSIONS/SIGNIFICANCE: The results have shown the capability of admixture to stimulate high-affinity and long-lasting antibodies against the target antigen to hinder Plasmodium parasite development in the mid-gut of An. stephensi. The attained results authenticated APN1/CMQ and APN1/CpG as a potent APN1-based TBV formulation which will be helpful in designing a vaccine in the future.


Subject(s)
Adjuvants, Immunologic , Anopheles , CD13 Antigens , Malaria Vaccines , Saponins , Animals , Anopheles/parasitology , Anopheles/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Mice , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Saponins/pharmacology , Saponins/administration & dosage , CD13 Antigens/immunology , CD13 Antigens/metabolism , Female , Plasmodium falciparum/immunology , Malaria/prevention & control , Malaria/transmission , Malaria/immunology , Malaria/parasitology , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Mice, Inbred BALB C , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology
2.
BMC Immunol ; 25(1): 44, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987710

ABSTRACT

BACKGROUND: Malaria is a life-threatening parasitic disease typically transmitted through the bite of an infected Anopheles mosquito. There is ample evidence showing the potential of malaria infection to affect the counts of lymphocyte subpopulations in the peripheral blood, but the extent of alteration might not be consistent in all geographical locations, due to several local factors. Although Ghana is among the malaria-endemic countries, there is currently no available data on the level of alterations that occur in the counts of lymphocyte subpopulations during P. falciparum malaria infection among adults. AIM: The study was to determine the immunophenotypic alterations in the level of peripheral blood lymphocytes and their subsets in adults with uncomplicated P. falciparum malaria infection and apparently healthy participants. METHODS: The study was a cross-sectional comparative study conducted in two municipalities of the Volta region of Ghana. Blood samples were collected from study participants and taken through serology (P. falciparum/Pan Rapid Diagnostic Kits), microscopy (Thick and thin blood films) and Haematological (Flow cytometric and Full blood count) analysis. RESULTS: A total of 414 participants, comprising 214 patients with malaria and 200 apparently healthy individuals (controls) were recruited into this study. Parasite density of the malaria patients ranged from 75/µL to 84,364/µL, with a mean of 3,520/µL. It was also observed that the total lymphocytes slightly decreased in the P. falciparum-infected individuals (Mean ± SD: 2.08 ± 4.93 × 109/L) compared to the control group (Mean ± SD: 2.47 ± 0.80 × 109/L). Again, there was a significant moderate positive correlation between parasite density and haematocrit levels (r = 0.321, p < 0.001). Apart from CD45 + T-cells, more people in the control group had normal values for the lymphocyte subsets measured compared to the malaria patients. CONCLUSIONS: From the results obtained, there was high parasite density among the malaria patients suggestive of high intensity of infection in the case group. The malaria patients again showed considerable haematological alterations in lymphocyte sub-sets and the parasite density appeared to be strongly associated with CD4 + T-cell reduction. Also, the parasite density significantly associated with decreasing haematocrit levels. This indicates that lymphocyte subset enumeration can be used to effectively support malaria diagnosis.


Subject(s)
Immunophenotyping , Malaria, Falciparum , Plasmodium falciparum , Humans , Malaria, Falciparum/immunology , Malaria, Falciparum/blood , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Male , Female , Adult , Plasmodium falciparum/immunology , Cross-Sectional Studies , Ghana , Middle Aged , Young Adult , Lymphocyte Subsets/immunology , Adolescent , Lymphocytes/immunology , Lymphocyte Count
4.
Front Immunol ; 15: 1358853, 2024.
Article in English | MEDLINE | ID: mdl-38835780

ABSTRACT

Introduction: Innate immunity is crucial to reducing parasite burden and contributing to survival in severe malaria. Monocytes are key actors in the innate response and, like macrophages, are plastic cells whose function and phenotype are regulated by the signals from the microenvironment. In the context of cerebral malaria (CM), monocyte response constitutes an important issue to understand. We previously demonstrated that decreased percentages of nonclassical monocytes were associated with death outcomes in CM children. In the current study, we postulated that monocyte phagocytosis function is impacted by the severity of malaria infection. Methods: To study this hypothesis, we compared the opsonic and nonopsonic phagocytosis capacity of circulant monocytes from Beninese children with uncomplicated malaria (UM) and CM. For the CM group, samples were obtained at inclusion (D0) and 3 and 30 days after treatment (D3, D30). The phagocytosis capacity of monocytes and their subsets was characterized by flow cytometry and transcriptional profiling by studying genes known for their functional implication in infected-red blood cell (iRBC) elimination or immune escape. Results: Our results confirm our hypothesis and highlight the higher capacity of nonclassical monocytes to phagocyte iRBC. We also confirm that a low number of nonclassical monocytes is associated with CM outcome when compared to UM, suggesting a mobilization of this subpopulation to the cerebral inflammatory site. Finally, our results suggest the implication of the inhibitory receptors LILRB1, LILRB2, and Tim3 in phagocytosis control. Discussion: Taken together, these data provide a better understanding of the interplay between monocytes and malaria infection in the pathogenicity of CM.


Subject(s)
Malaria, Cerebral , Monocytes , Phagocytosis , Humans , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Monocytes/immunology , Male , Child, Preschool , Female , Child , Infant , Plasmodium falciparum/immunology , Opsonin Proteins/metabolism , Opsonin Proteins/immunology , Erythrocytes/parasitology , Erythrocytes/immunology , Immunity, Innate
5.
Nat Commun ; 15(1): 4913, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851821

ABSTRACT

Host immune responses are tightly controlled by various immune factors during infection, and protozoan parasites also manipulate the immune system to evade surveillance, leading to an evolutionary arms race in host‒pathogen interactions; however, the underlying mechanisms are not fully understood. We observed that the level of superoxide dismutase 3 (SOD3) was significantly elevated in both Plasmodium falciparum malaria patients and mice infected with four parasite species. SOD3-deficient mice had a substantially longer survival time and lower parasitemia than control mice after infection, whereas SOD3-overexpressing mice were much more vulnerable to parasite infection. We revealed that SOD3, secreted from activated neutrophils, bound to T cells, suppressed the interleukin-2 expression and concomitant interferon-gamma responses crucial for parasite clearance. Overall, our findings expose active fronts in the arms race between the parasites and host immune system and provide insights into the roles of SOD3 in shaping host innate immune responses to parasite infection.


Subject(s)
Malaria, Falciparum , Mice, Inbred C57BL , Mice, Knockout , Neutrophils , Superoxide Dismutase , Animals , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics , Humans , Mice , Neutrophils/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Immunity, Cellular , T-Lymphocytes/immunology , Plasmodium falciparum/immunology , Female , Host-Parasite Interactions/immunology , Host-Parasite Interactions/genetics , Interferon-gamma/metabolism , Interferon-gamma/immunology , Male , Immunity, Innate , Interleukin-2/metabolism , Interleukin-2/immunology , Interleukin-2/genetics , Parasitemia/immunology
6.
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
7.
Nat Commun ; 15(1): 4857, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849365

ABSTRACT

Reticulocyte-binding protein homologue 5 (RH5), a leading blood-stage Plasmodium falciparum malaria vaccine target, interacts with cysteine-rich protective antigen (CyRPA) and RH5-interacting protein (RIPR) to form an essential heterotrimeric "RCR-complex". We investigate whether RCR-complex vaccination can improve upon RH5 alone. Using monoclonal antibodies (mAbs) we show that parasite growth-inhibitory epitopes on each antigen are surface-exposed on the RCR-complex and that mAb pairs targeting different antigens can function additively or synergistically. However, immunisation of female rats with the RCR-complex fails to outperform RH5 alone due to immuno-dominance of RIPR coupled with inferior potency of anti-RIPR polyclonal IgG. We identify that all growth-inhibitory antibody epitopes of RIPR cluster within the C-terminal EGF-like domains and that a fusion of these domains to CyRPA, called "R78C", combined with RH5, improves the level of in vitro parasite growth inhibition compared to RH5 alone. These preclinical data justify the advancement of the RH5.1 + R78C/Matrix-M™ vaccine candidate to Phase 1 clinical trial.


Subject(s)
Antibodies, Monoclonal , Antibodies, Protozoan , Antigens, Protozoan , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Animals , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Female , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Antigens, Protozoan/immunology , Rats , Antibodies, Protozoan/immunology , Antibodies, Monoclonal/immunology , Humans , Epitopes/immunology , Carrier Proteins/immunology , Carrier Proteins/metabolism
8.
PLoS Comput Biol ; 20(6): e1012131, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38848436

ABSTRACT

Immunization through repeated direct venous inoculation of Plasmodium falciparum (Pf) sporozoites (PfSPZ) under chloroquine chemoprophylaxis, using the PfSPZ Chemoprophylaxis Vaccine (PfSPZ-CVac), induces high-level protection against controlled human malaria infection (CHMI). Humoral and cellular immunity contribute to vaccine efficacy but only limited information about the implicated Pf-specific antigens is available. Here, we examined Pf-specific antibody profiles, measured by protein arrays representing the full Pf proteome, of 40 placebo- and PfSPZ-immunized malaria-naïve volunteers from an earlier published PfSPZ-CVac dose-escalation trial. For this purpose, we both utilized and adapted supervised machine learning methods to identify predictive antibody profiles at two different time points: after immunization and before CHMI. We developed an adapted multitask support vector machine (SVM) approach and compared it to standard methods, i.e. single-task SVM, regularized logistic regression and random forests. Our results show, that the multitask SVM approach improved the classification performance to discriminate the protection status based on the underlying antibody-profiles while combining time- and dose-dependent data in the prediction model. Additionally, we developed the new fEature diStance exPlainabilitY (ESPY) method to quantify the impact of single antigens on the non-linear multitask SVM model and make it more interpretable. In conclusion, our multitask SVM model outperforms the studied standard approaches in regard of classification performance. Moreover, with our new explanation method ESPY, we were able to interpret the impact of Pf-specific antigen antibody responses that predict sterile protective immunity against CHMI after immunization. The identified Pf-specific antigens may contribute to a better understanding of immunity against human malaria and may foster vaccine development.


Subject(s)
Antibodies, Protozoan , Machine Learning , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Malaria Vaccines/immunology , Humans , Plasmodium falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Vaccine Efficacy , Support Vector Machine , Computational Biology/methods
9.
Nat Commun ; 15(1): 5194, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890271

ABSTRACT

Resistance to clinical malaria takes years to develop even in hyperendemic regions and sterilizing immunity has rarely been observed. To evaluate the maturation of the host response against controlled repeat exposures to P. falciparum (Pf) NF54 strain-infected mosquitoes, we systematically monitored malaria-naïve participants through an initial exposure to uninfected mosquitoes and 4 subsequent homologous exposures to Pf-infected mosquitoes over 21 months (n = 8 males) (ClinicalTrials.gov# NCT03014258). The primary outcome was to determine whether protective immunity against parasite infection develops following repeat CHMI and the secondary outcomes were to track the clinical signs and symptoms of malaria and anti-Pf antibody development following repeat CHMI. After two exposures, time to blood stage patency increases significantly and the number of reported symptoms decreases indicating the development of clinical tolerance. The time to patency correlates positively with both anti-Pf circumsporozoite protein (CSP) IgG and CD8 + CD69+ effector memory T cell levels consistent with partial pre-erythrocytic immunity. IFNγ levels decrease significantly during the participants' second exposure to high blood stage parasitemia and could contribute to the decrease in symptoms. In contrast, CD4-CD8 + T cells expressing CXCR5 and the inhibitory receptor, PD-1, increase significantly after subsequent Pf exposures, possibly dampening the memory response and interfering with the generation of robust sterilizing immunity.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Humans , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/blood , Plasmodium falciparum/immunology , Male , Protozoan Proteins/immunology , Animals , Adult , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Interferon-gamma/metabolism , Interferon-gamma/immunology , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Young Adult , CD8-Positive T-Lymphocytes/immunology , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Anopheles/parasitology
10.
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
11.
Front Immunol ; 15: 1385380, 2024.
Article in English | MEDLINE | ID: mdl-38827744

ABSTRACT

Introduction: Depending on the microenvironment, γδ T cells may assume characteristics similar to those of Th1, Th2, Th17, regulatory T cells or antigen presenting cells. Despite the wide documentation of the effect of Th1/Th2 balance on pregnancy associated malaria and outcomes, there are no reports on the relationship between γδ T cell phenotype change and Placental Malaria (PM) with pregnancy outcomes. This study sought to investigate the involvement of γδ T cells and its subsets in placental Plasmodium falciparum malaria. Methods: In a case-control study conducted in Yaoundé, Cameroon from March 2022 to May 2023, peripheral, placental and cord blood samples were collected from 50 women at delivery (29 PM negative: PM- and 21 PM positive: PM+; as diagnosed by light microscopy). Hemoglobin levels were measured using hemoglobinometer. PBMCs, IVBMCs and CBMCs were isolated using histopaque-1077 and used to characterize total γδ T cell populations and subsets (Vδ1+, Vδ2+, Vδ1-Vδ2-) by flow cytometry. Results: Placental Plasmodium falciparum infection was associated with significant increase in the frequency of total γδ T cells in IVBMC and of the Vδ1+ subset in PBMC and IVBMC, but decreased frequency of the Vδ2+ subset in PBMC and IVBMC. The expression of the activation marker: HLA-DR, and the exhaustion markers (PD1 and TIM3) within total γδ T cells and subsets were significantly up-regulated in PM+ compared to PM- group. The frequency of total γδ T cells in IVBMC, TIM-3 expression within total γδ T cells and subsets in IVBMC, as well as HLA-DR expression within total γδ T cells and Vδ2+ subset in IVBMC were negatively associated with maternal hemoglobin levels. Furthermore, the frequency of total γδ T cells in PBMC and PD1 expression within the Vδ2+ subset in CBMC were negatively associated with birth weight contrary to the frequency of Vδ1-Vδ2- subset in PBMC and HLA-DR expression within the Vδ2+ subset in IVBMC which positively associated with maternal hemoglobin level and birth weight, respectively. Conclusion: The data indicate up-regulation of activated and exhausted γδ T cells in Plasmodium falciparum placental malaria, with effects on pregnancy outcomes including maternal hemoglobin level and birth weight.


Subject(s)
Malaria, Falciparum , Placenta , Plasmodium falciparum , Pregnancy Complications, Parasitic , Pregnancy Outcome , Receptors, Antigen, T-Cell, gamma-delta , Humans , Female , Pregnancy , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/blood , Cameroon , Adult , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Case-Control Studies , Young Adult , Placenta/immunology , Placenta/parasitology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Phenotype
12.
Front Cell Infect Microbiol ; 14: 1375249, 2024.
Article in English | MEDLINE | ID: mdl-38808064

ABSTRACT

Introduction: Diversity in malarial antigens is an immune evasion mechanism that gives malaria parasites an edge over the host. Immune responses against one variant of a polymorphic antigen are usually not fully effective against other variants due to altered epitopes. This study aimed to evaluate diversity in the Plasmodium falciparum antigens apical membrane antigen 1 (PfAMA1) and circumsporozoite protein (PfCSP) from circulating parasites in a malaria-endemic community in southern Ghana and to determine the effects of polymorphisms on antibody response specificity. Methods: The study involved 300 subjects, whose P. falciparum infection status was determined by microscopy and PCR. Diversity within the two antigens was evaluated by msp2 gene typing and molecular gene sequencing, while the host plasma levels of antibodies against PfAMA1, PfCSP, and two synthetic 24mer peptides from the conserved central repeat region of PfCSP, were measured by ELISA. Results: Of the 300 subjects, 171 (57%) had P. falciparum infection, with 165 of the 171 (96.5%) being positive for either or both of the msp2 allelic families. Gene sequencing of DNA from 55 clonally infected samples identified a total of 56 non-synonymous single nucleotide polymorphisms (SNPs) for the Pfama1 gene and these resulted in 44 polymorphic positions, including two novel positions (363 and 365). Sequencing of the Pfcsp gene from 69 clonal DNA samples identified 50 non-synonymous SNPs that resulted in 42 polymorphic positions, with half (21) of these polymorphic positions being novel. Of the measured antibodies, only anti-PfCSP antibodies varied considerably between PCR parasite-positive and parasite-negative persons. Discussion: These data confirm the presence of a considerable amount of unique, previously unreported amino acid changes, especially within PfCSP. Drivers for this diversity in the Pfcsp gene do not immediately seem apparent, as immune pressure will be expected to drive a similar level of diversity in the Pfama1 gene.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Malaria, Falciparum , Membrane Proteins , Plasmodium falciparum , Protozoan Proteins , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Ghana , Humans , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Female , Adult , Male , Adolescent , Young Adult , Child , Genetic Variation , Child, Preschool , Middle Aged , Sequence Analysis, DNA , Enzyme-Linked Immunosorbent Assay , Polymerase Chain Reaction , Antigenic Variation , DNA, Protozoan/genetics
13.
Malar J ; 23(1): 154, 2024 May 19.
Article in English | MEDLINE | ID: mdl-38764069

ABSTRACT

BACKGROUND: Plasmodium falciparum malaria is a public health issue mostly seen in tropical countries. Until now, there is no effective malaria vaccine against antigens specific to the blood-stage of P. falciparum infection. Because the pathogenesis of malarial disease results from blood-stage infection, it is essential to identify the most promising blood-stage vaccine candidate antigens under natural exposure to malaria infection. METHODS: A cohort of 400 pregnant women and their infants was implemented in South Benin. An active and passive protocol of malaria surveillance was established during pregnancy and infancy to precisely ascertain malaria infections during the follow-up. Twenty-eight antibody (Ab) responses specific to seven malaria candidate vaccine antigens were repeatedly quantified during pregnancy (3 time points) and infancy (6 time points) in order to study the Ab kinetics and their protective role. Abs were quantified by ELISA and logistic, linear and cox-proportional hazard model were performed to analyse the associations between Ab responses and protection against malaria in mothers and infants, taking into account socio-economic factors and for infants an environmental risk of exposure. RESULTS: The levels of IgM against MSP1, MSP2 and MSP3 showed an early protective response against the onset of symptomatic malaria infections starting from the 18th month of life, whereas no association was found for IgG responses during infancy. In women, some IgG responses tend to be associated with a protection against malaria risk along pregnancy and at delivery, among them IgG3 against GLURP-R0 and IgG2 against MSP1. CONCLUSION: The main finding suggests that IgM should be considered in vaccine designs during infanthood. Investigation of the functional role played by IgM in malaria protection needs further attention.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Immunoglobulin G , Immunoglobulin M , Malaria, Falciparum , Plasmodium falciparum , Humans , Female , Plasmodium falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Pregnancy , Infant , Immunoglobulin M/blood , Immunoglobulin G/blood , Antibodies, Protozoan/blood , Benin , Antigens, Protozoan/immunology , Adult , Young Adult , Enzyme-Linked Immunosorbent Assay , Infant, Newborn , Pregnancy Complications, Parasitic/prevention & control , Pregnancy Complications, Parasitic/immunology , Cohort Studies
14.
Biochem Soc Trans ; 52(3): 1025-1034, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38752830

ABSTRACT

Despite having the highest risk of progressing to severe disease due to lack of acquired immunity, the youngest children living in areas of highly intense malaria transmission have long been observed to be infected at lower rates than older children. Whether this observation is due to reduced exposure to infectious mosquito bites from behavioral and biological factors, maternally transferred immunity, genetic factors, or enhanced innate immunity in the young child has intrigued malaria researchers for over half a century. Recent evidence suggests that maternally transferred immunity may be limited to early infancy and that the young child's own immune system may contribute to control of malarial symptoms early in life and prior to the development of more effective adaptive immunity. Prospective studies of active and passive detection of Plasmodium falciparum blood-stage infections have identified young children (<5 years old) who remain uninfected through a defined surveillance period despite living in settings of highly intense malaria transmission. Yet, little is known about the potential immunological basis for this 'aparasitemic' phenotype. In this review, we summarize the observational evidence for this phenotype in field studies and examine potential reasons why these children escape detection of parasitemia, covering factors that are either extrinsic or intrinsic to their developing immune system. We discuss the challenges of distinguishing malaria protection from lack of malaria exposure in field studies. We also identify gaps in our knowledge regarding cellular immunity in the youngest age group and propose directions that researchers may take to address these gaps.


Subject(s)
Malaria, Falciparum , Parasitemia , Plasmodium falciparum , Humans , Child, Preschool , Malaria, Falciparum/transmission , Plasmodium falciparum/immunology , Infant , Malaria/transmission , Immunity, Innate , Animals
15.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38803222

ABSTRACT

The merozoite surface protein 1 (MSP1) is the most abundant protein on the surface of the invasive merozoite stages of Plasmodium falciparum and has long been considered a key target of protective immunity. We used samples from a single controlled human malaria challenge study to test whether the full-length version of MSP1 (MSP1FL) induced antibodies that mediated Fc-IgG functional activity in five independent assays. We found that anti-MSP1FL antibodies induced complement fixation via C1q, monocyte-mediated phagocytosis, neutrophil respiratory burst, and natural killer cell degranulation as well as IFNγ production. Activity in each of these assays was strongly associated with protection. The breadth of MSP1-specific Fc-mediated effector functions was more strongly associated with protection than the individual measures and closely mirrored what we have previously reported using the same assays against merozoites. Our findings suggest that MSP1FL is an important target of functional antibodies that contribute to a protective immune response against malaria.


Subject(s)
Antibodies, Protozoan , Malaria, Falciparum , Merozoite Surface Protein 1 , Phagocytosis , Plasmodium falciparum , Humans , Merozoite Surface Protein 1/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Antibodies, Protozoan/immunology , Phagocytosis/immunology , Immunoglobulin G/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Female , Merozoites/immunology , Neutrophils/immunology , Neutrophils/metabolism
16.
Immunity ; 57(6): 1215-1224.e6, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38788711

ABSTRACT

Malaria is a life-threatening disease of global health importance, particularly in sub-Saharan Africa. The growth inhibition assay (GIA) is routinely used to evaluate, prioritize, and quantify the efficacy of malaria blood-stage vaccine candidates but does not reliably predict either naturally acquired or vaccine-induced protection. Controlled human malaria challenge studies in semi-immune volunteers provide an unparalleled opportunity to robustly identify mechanistic correlates of protection. We leveraged this platform to undertake a head-to-head comparison of seven functional antibody assays that are relevant to immunity against the erythrocytic merozoite stage of Plasmodium falciparum. Fc-mediated effector functions were strongly associated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following the challenge. These findings present a shift in the understanding of the mechanisms that underpin immunity to malaria and have important implications for vaccine development.


Subject(s)
Antibodies, Protozoan , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Humans , Plasmodium falciparum/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Adult , Immunoglobulin Fc Fragments/immunology , Merozoites/immunology , Erythrocytes/parasitology , Erythrocytes/immunology , Female , Male , Young Adult
17.
JCI Insight ; 9(9)2024 May 08.
Article in English | MEDLINE | ID: mdl-38716733

ABSTRACT

Vaccination of malaria-naive volunteers with a high dose of Plasmodium falciparum sporozoites chemoattenuated by chloroquine (CQ) (PfSPZ-CVac [CQ]) has previously demonstrated full protection against controlled human malaria infection (CHMI). However, lower doses of PfSPZ-CVac [CQ] resulted in incomplete protection. This provides the opportunity to understand the immune mechanisms needed for better vaccine-induced protection by comparing individuals who were protected with those not protected. Using mass cytometry, we characterized immune cell composition and responses of malaria-naive European volunteers who received either lower doses of PfSPZ-CVac [CQ], resulting in 50% protection irrespective of the dose, or a placebo vaccination, with everyone becoming infected following CHMI. Clusters of CD4+ and γδ T cells associated with protection were identified, consistent with their known role in malaria immunity. Additionally, EMRA CD8+ T cells and CD56+CD8+ T cell clusters were associated with protection. In a cohort from a malaria-endemic area in Gabon, these CD8+ T cell clusters were also associated with parasitemia control in individuals with lifelong exposure to malaria. Upon stimulation with P. falciparum-infected erythrocytes, CD4+, γδ, and EMRA CD8+ T cells produced IFN-γ and/or TNF, indicating their ability to mediate responses that eliminate malaria parasites.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Sporozoites , Adolescent , Adult , Female , Humans , Male , Young Adult , Antimalarials/therapeutic use , Antimalarials/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chloroquine/therapeutic use , Chloroquine/pharmacology , Europe , European People , Gabon , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Parasitemia/immunology , Plasmodium falciparum/immunology , Sporozoites/immunology , Vaccination/methods , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Central African People
18.
PLoS Pathog ; 20(4): e1012145, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38598552

ABSTRACT

Wolbachia, a maternally transmitted symbiotic bacterium of insects, can suppress a variety of human pathogens in mosquitoes, including malaria-causing Plasmodium in the Anopheles vector. However, the mechanistic basis of Wolbachia-mediated Plasmodium suppression in mosquitoes is not well understood. In this study, we compared the midgut and carcass transcriptomes of stably infected Anopheles stephensi with Wolbachia wAlbB to uninfected mosquitoes in order to discover Wolbachia infection-responsive immune genes that may play a role in Wolbachia-mediated anti-Plasmodium activity. We show that wAlbB infection upregulates 10 putative immune genes and downregulates 14 in midguts, while it upregulates 31 putative immune genes and downregulates 15 in carcasses at 24 h after blood-fed feeding, the time at which the Plasmodium ookinetes are traversing the midgut tissue. Only a few of these regulated immune genes were also significantly differentially expressed between Wolbachia-infected and non-infected midguts and carcasses of sugar-fed mosquitoes. Silencing of the Wolbachia infection-responsive immune genes TEP 4, TEP 15, lysozyme C2, CLIPB2, CLIPB4, PGRP-LD and two novel genes (a peritrophin-44-like gene and a macro domain-encoding gene) resulted in a significantly greater permissiveness to P. falciparum infection. These results indicate that Wolbachia infection modulates mosquito immunity and other processes that are likely to decrease Anopheles permissiveness to Plasmodium infection.


Subject(s)
Anopheles , Malaria, Falciparum , Plasmodium falciparum , Wolbachia , Animals , Anopheles/parasitology , Anopheles/microbiology , Anopheles/immunology , Wolbachia/immunology , Plasmodium falciparum/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mosquito Vectors/parasitology , Mosquito Vectors/microbiology , Mosquito Vectors/immunology , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Proteins/immunology , Transcriptome , Female
19.
NPJ Syst Biol Appl ; 10(1): 44, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678051

ABSTRACT

Malaria vaccine development is hampered by extensive antigenic variation and complex life stages of Plasmodium species. Vaccine development has focused on a small number of antigens, many of which were identified without utilizing systematic genome-level approaches. In this study, we implement a machine learning-based reverse vaccinology approach to predict potential new malaria vaccine candidate antigens. We assemble and analyze P. falciparum proteomic, structural, functional, immunological, genomic, and transcriptomic data, and use positive-unlabeled learning to predict potential antigens based on the properties of known antigens and remaining proteins. We prioritize candidate antigens based on model performance on reference antigens with different genetic diversity and quantify the protein properties that contribute most to identifying top candidates. Candidate antigens are characterized by gene essentiality, gene ontology, and gene expression in different life stages to inform future vaccine development. This approach provides a framework for identifying and prioritizing candidate vaccine antigens for a broad range of pathogens.


Subject(s)
Antigens, Protozoan , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Plasmodium falciparum/immunology , Plasmodium falciparum/genetics , Malaria Vaccines/immunology , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Machine Learning , Humans , Proteomics/methods , Vaccine Development/methods , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Computational Biology/methods
20.
BMC Med ; 22(1): 170, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38649867

ABSTRACT

BACKGROUND: The stalling global progress in malaria control highlights the need for novel tools for malaria elimination, including transmission-blocking vaccines. Transmission-blocking vaccines aim to induce human antibodies that block parasite development in the mosquito and mosquitoes becoming infectious. The Pfs48/45 protein is a leading Plasmodium falciparum transmission-blocking vaccine candidate. The R0.6C fusion protein, consisting of Pfs48/45 domain 3 (6C) and the N-terminal region of P. falciparum glutamate-rich protein (R0), has previously been produced in Lactococcus lactis and elicited functional antibodies in rodents. Here, we assess the safety and transmission-reducing efficacy of R0.6C adsorbed to aluminium hydroxide with and without Matrix-M™ adjuvant in humans. METHODS: In this first-in-human, open-label clinical trial, malaria-naïve adults, aged 18-55 years, were recruited at the Radboudumc in Nijmegen, the Netherlands. Participants received four intramuscular vaccinations on days 0, 28, 56 and 168 with either 30 µg or 100 µg of R0.6C and were randomised for the allocation of one of the two different adjuvant combinations: aluminium hydroxide alone, or aluminium hydroxide combined with Matrix-M1™ adjuvant. Adverse events were recorded from inclusion until 84 days after the fourth vaccination. Anti-R0.6C and anti-6C IgG titres were measured by enzyme-linked immunosorbent assay. Transmission-reducing activity of participants' serum and purified vaccine-specific immunoglobulin G was assessed by standard membrane feeding assays using laboratory-reared Anopheles stephensi mosquitoes and cultured P. falciparum gametocytes. RESULTS: Thirty-one participants completed four vaccinations and were included in the analysis. Administration of all doses was safe and well-tolerated, with one related grade 3 adverse event (transient fever) and no serious adverse events occurring. Anti-R0.6C and anti-6C IgG titres were similar between the 30 and 100 µg R0.6C arms, but higher in Matrix-M1™ arms. Neat participant sera did not induce significant transmission-reducing activity in mosquito feeding experiments, but concentrated vaccine-specific IgGs purified from sera collected two weeks after the fourth vaccination achieved up to 99% transmission-reducing activity. CONCLUSIONS: R0.6C/aluminium hydroxide with or without Matrix-M1™ is safe, immunogenic and induces functional Pfs48/45-specific transmission-blocking antibodies, albeit at insufficient serum concentrations to result in transmission reduction by neat serum. Future work should focus on identifying alternative vaccine formulations or regimens that enhance functional antibody responses. TRIAL REGISTRATION: The trial is registered with ClinicalTrials.gov under identifier NCT04862416.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Membrane Glycoproteins , Plasmodium falciparum , Protozoan Proteins , Adolescent , Adult , Animals , Female , Humans , Male , Middle Aged , Young Adult , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Antibodies, Protozoan , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Falciparum/immunology , Netherlands , Plasmodium falciparum/immunology , Protozoan Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...