Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
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
2.
Front Immunol ; 15: 1360220, 2024.
Article in English | MEDLINE | ID: mdl-38650925

ABSTRACT

Background: Malaria remains a major global health priority, and monoclonal antibodies (mAbs) are emerging as potential new tools to support efforts to control the disease. Recent data suggest that Fc-dependent mechanisms of immunity are important mediators of protection against the blood stages of the infection, but few studies have investigated this in the context of mAbs. We aimed to isolate mAbs agnostic to cognate antigens that target whole merozoites and simultaneously induce potent neutrophil activity measured by the level of reactive oxygen species (ROS) production using an antibody-dependent respiratory burst (ADRB) assay. Methods: We used samples from semi-immune adults living in coastal Kenya to isolate mAbs that induce merozoite-specific ADRB activity. We then tested whether modifying the expressed IgG1 isotype to an IgG-IgA Fc region chimera would enhance the level of ADRB activity. Results: We isolated a panel of nine mAbs with specificity to whole merozoites. mAb J31 induced ADRB activity in a dose-dependent fashion. Compared to IgG1, our modified antibody IgG-IgA bi-isotype induced higher ADRB activity across all concentrations tested. Further, we observed a negative hook effect at high IgG1 mAb concentrations (i.e., >200 µg/mL), but this was reversed by Fc modification. We identified MSP3.5 as the potential cognate target of mAb J31. Conclusions: We demonstrate an approach to engineer mAbs with enhanced ADRB potency against blood-stage parasites.


Subject(s)
Antibodies, Monoclonal , Antibodies, Protozoan , Malaria, Falciparum , Merozoites , Neutrophils , Plasmodium falciparum , Plasmodium falciparum/immunology , Humans , Antibodies, Protozoan/immunology , Neutrophils/immunology , Neutrophils/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Antibodies, Monoclonal/immunology , Merozoites/immunology , Respiratory Burst/immunology , Immunoglobulin G/immunology , Adult , Reactive Oxygen Species/metabolism , Kenya , Immunoglobulin Isotypes/immunology , Neutrophil Activation/immunology , Female , Antigens, Protozoan/immunology
3.
Sci Transl Med ; 15(682): eabn5993, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36753561

ABSTRACT

Natural killer (NK) cells are potent immune effectors that can be activated via antibody-mediated Fc receptor engagement. Using multiparameter flow cytometry, we found that NK cells degranulate and release IFN-γ upon stimulation with antibody-opsonized Plasmodium falciparum merozoites. Antibody-dependent NK (Ab-NK) activity was largely strain transcending and enhanced invasion inhibition into erythrocytes. Ab-NK was associated with the successful control of parasitemia after experimental malaria challenge in African adults. In an independent cohort study in children, Ab-NK increased with age, was boosted by concurrent P. falciparum infections, and was associated with a lower risk of clinical episodes of malaria. Nine of the 14 vaccine candidates tested induced Ab-NK, including some less well-characterized antigens: P41, P113, MSP11, RHOPH3, and Pf_11363200. These data highlight an important role of Ab-NK activity in immunity against malaria and provide a potential mechanism for evaluating vaccine candidates.


Subject(s)
Malaria, Falciparum , Malaria , Child , Adult , Animals , Humans , Antigens, Protozoan , Cohort Studies , Merozoites , Antibodies, Protozoan , Plasmodium falciparum , Killer Cells, Natural
4.
Nat Commun ; 13(1): 331, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039519

ABSTRACT

Strengthening malaria surveillance is a key intervention needed to reduce the global disease burden. Reliable serological markers of recent malaria exposure could improve current surveillance methods by allowing for accurate estimates of infection incidence from limited data. We studied the IgG antibody response to 111 Plasmodium falciparum proteins in 65 adult travellers followed longitudinally after a natural malaria infection in complete absence of re-exposure. We identified a combination of five serological markers that detect exposure within the previous three months with >80% sensitivity and specificity. Using mathematical modelling, we examined the antibody kinetics and determined that responses informative of recent exposure display several distinct characteristics: rapid initial boosting and decay, less inter-individual variation in response kinetics, and minimal persistence over time. Such serological exposure markers could be incorporated into routine malaria surveillance to guide efforts for malaria control and elimination.


Subject(s)
Antibodies, Protozoan/immunology , Biomarkers/metabolism , Malaria/epidemiology , Malaria/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adult , Antibody Formation/immunology , Child , Child, Preschool , Cohort Studies , Epitopes/immunology , Female , Fluorescence , Humans , Infant , Kenya/epidemiology , Kinetics , Male , Middle Aged , Models, Biological , ROC Curve , Young Adult
5.
Trends Parasitol ; 37(4): 265-267, 2021 04.
Article in English | MEDLINE | ID: mdl-33518435

ABSTRACT

The path to a successful international career in science is daunting for most, and women in Africa are no exception. We are grossly under-represented as senior authors in high-impact publications and keynote speakers at international conferences, and rarely head major funding networks. Smashing long-held stereotypes may ease the journey.


Subject(s)
Science , Africa , Cultural Diversity , Female , Humans , Publications/statistics & numerical data , Respect , Science/statistics & numerical data , Sex Factors , Stereotyping
6.
Methods Mol Biol ; 2013: 83-90, 2019.
Article in English | MEDLINE | ID: mdl-31267495

ABSTRACT

The enzyme-linked immunosorbent assay (ELISA) is a reliable and relatively low-cost method for measuring soluble ligands such as antibodies and proteins in biological samples. For analysis of specific antibodies in serum, a capture antigen is immobilized onto a solid polystyrene surface from which it can capture the antibodies. The captured antibodies are subsequently detected using a secondary antibody conjugated to an enzyme. Detection is accomplished by addition of a colorimetric substrate, and the readout is absorbance (optical density). Here, we provide a detailed standardized ELISA protocol for the quantification of antibodies against malaria antigens.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Antibodies/analysis , Antibodies/immunology , Antigens/analysis , Antigens/immunology , Antimalarials/therapeutic use , Humans , Malaria/diagnosis , Malaria/immunology
7.
Front Immunol ; 9: 2866, 2018.
Article in English | MEDLINE | ID: mdl-30619257

ABSTRACT

Passive transfer studies in humans clearly demonstrated the protective role of IgG antibodies against malaria. Identifying the precise parasite antigens that mediate immunity is essential for vaccine design, but has proved difficult. Completion of the Plasmodium falciparum genome revealed thousands of potential vaccine candidates, but a significant bottleneck remains in their validation and prioritization for further evaluation in clinical trials. Focusing initially on the Plasmodium falciparum merozoite proteome, we used peer-reviewed publications, multiple proteomic and bioinformatic approaches, to select and prioritize potential immune targets. We expressed 109 P. falciparum recombinant proteins, the majority of which were obtained using a mammalian expression system that has been shown to produce biologically functional extracellular proteins, and used them to create KILchip v1.0: a novel protein microarray to facilitate high-throughput multiplexed antibody detection from individual samples. The microarray assay was highly specific; antibodies against P. falciparum proteins were detected exclusively in sera from malaria-exposed but not malaria-naïve individuals. The intensity of antibody reactivity varied as expected from strong to weak across well-studied antigens such as AMA1 and RH5 (Kruskal-Wallis H test for trend: p < 0.0001). The inter-assay and intra-assay variability was minimal, with reproducible results obtained in re-assays using the same chip over a duration of 3 months. Antibodies quantified using the multiplexed format in KILchip v1.0 were highly correlated with those measured in the gold-standard monoplex ELISA [median (range) Spearman's R of 0.84 (0.65-0.95)]. KILchip v1.0 is a robust, scalable and adaptable protein microarray that has broad applicability to studies of naturally acquired immunity against malaria by providing a standardized tool for the detection of antibody correlates of protection. It will facilitate rapid high-throughput validation and prioritization of potential Plasmodium falciparum merozoite-stage antigens paving the way for urgently needed clinical trials for the next generation of malaria vaccines.


Subject(s)
Malaria, Falciparum/immunology , Merozoites/immunology , Plasmodium falciparum/immunology , Protein Array Analysis/methods , Proteome/immunology , Proteomics/methods , Health Priorities , Malaria Vaccines/immunology , Malaria, Falciparum/microbiology , Merozoites/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Proteome/metabolism , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Research
8.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093263

ABSTRACT

The development of a highly effective vaccine remains a key strategic goal to aid the control and eventual eradication of Plasmodium falciparum malaria. In recent years, the reticulocyte-binding protein homolog 5 (RH5) has emerged as the most promising blood-stage P. falciparum candidate antigen to date, capable of conferring protection against stringent challenge in Aotus monkeys. We report on the first clinical trial to our knowledge to assess the RH5 antigen - a dose-escalation phase Ia study in 24 healthy, malaria-naive adult volunteers. We utilized established viral vectors, the replication-deficient chimpanzee adenovirus serotype 63 (ChAd63), and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA), encoding RH5 from the 3D7 clone of P. falciparum. Vaccines were administered i.m. in a heterologous prime-boost regimen using an 8-week interval and were well tolerated. Vaccine-induced anti-RH5 serum antibodies exhibited cross-strain functional growth inhibition activity (GIA) in vitro, targeted linear and conformational epitopes within RH5, and inhibited key interactions within the RH5 invasion complex. This is the first time to our knowledge that substantial RH5-specific responses have been induced by immunization in humans, with levels greatly exceeding the serum antibody responses observed in African adults following years of natural malaria exposure. These data support the progression of RH5-based vaccines to human efficacy testing.


Subject(s)
Antibodies, Neutralizing , Carrier Proteins/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Vaccination , Adaptive Immunity , Adult , Antibodies, Protozoan/blood , Carrier Proteins/genetics , Epitopes/immunology , Female , Genetic Vectors , Humans , Immunization , Male , Middle Aged , Plasmodium falciparum/genetics , Vaccinia virus , Young Adult
9.
Immunology ; 152(2): 195-206, 2017 10.
Article in English | MEDLINE | ID: mdl-28646586

ABSTRACT

Although epidemiological observations, IgG passive transfer studies and experimental infections in humans all support the feasibility of developing highly effective malaria vaccines, the precise antigens that induce protective immunity remain uncertain. Here, we review the methodologies applied to vaccine candidate discovery for Plasmodium falciparum malaria from the pre- to post-genomic era. Probing of genomic and cDNA libraries with antibodies of defined specificities or functional activity predominated the former, whereas reverse vaccinology encompassing high throughput in silico analyses of genomic, transcriptomic or proteomic parasite data sets is the mainstay of the latter. Antibody-guided vaccine design spanned both eras but currently benefits from technological advances facilitating high-throughput screening and downstream applications. We make the case that although we have exponentially increased our ability to identify numerous potential vaccine candidates in a relatively short space of time, a significant bottleneck remains in their validation and prioritization for evaluation in clinical trials. Longitudinal cohort studies provide supportive evidence but results are often conflicting between studies. Demonstration of antigen-specific antibody function is valuable but the relative importance of one mechanism over another with regards to protection remains undetermined. Animal models offer useful insights but may not accurately reflect human disease. Challenge studies in humans are preferable but prohibitively expensive. In the absence of reliable correlates of protection, suitable animal models or a better understanding of the mechanisms underlying protective immunity in humans, vaccine candidate discovery per se may not be sufficient to provide the paradigm shift necessary to develop the next generation of highly effective subunit malaria vaccines.


Subject(s)
Antigens, Protozoan/immunology , Drug Discovery/methods , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Gene Library , High-Throughput Screening Assays , Host-Pathogen Interactions , Humans , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Proteomics
10.
Int J Parasitol ; 47(2-3): 153-162, 2017 02.
Article in English | MEDLINE | ID: mdl-27890694

ABSTRACT

Young infants are less susceptible to severe episodes of malaria but the targets and mechanisms of protection are not clear. Cord blood antibodies may play an important role in mediating protection but many studies have examined their association with the outcome of infection or non-severe malaria. Here, we investigated whether cord blood IgG to Plasmodium falciparum merozoite antigens and antibody-mediated effector functions were associated with reduced odds of developing severe malaria at different time points during the first year of life. We conducted a case-control study of well-defined severe falciparum malaria nested within a longitudinal birth cohort of Kenyan children. We measured cord blood total IgG levels against five recombinant merozoite antigens and antibody function in the growth inhibition activity and neutrophil antibody-dependent respiratory burst assays. We also assessed the decay of maternal antibodies during the first 6months of life. The mean antibody half-life range was 2.51months (95% confidence interval (CI): 2.19-2.92) to 4.91months (95% CI: 4.47-6.07). The rate of decline of maternal antibodies was inversely proportional to the starting concentration. The functional assay of antibody-dependent respiratory burst activity predicted significantly reduced odds of developing severe malaria during the first 6months of life (Odds ratio (OR) 0.07, 95% CI: 0.007-0.74, P=0.007). Identification of the targets of antibodies mediating antibody-dependent respiratory burst activity could contribute to the development of malaria vaccines that protect against severe episodes of malaria in early infancy.


Subject(s)
Fetal Blood/immunology , Immunoglobulin G/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Case-Control Studies , Cohort Studies , Female , Humans , Infant , Kenya , Male , Respiratory Burst , Risk Factors
11.
Infect Immun ; 84(4): 950-963, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26787721

ABSTRACT

Severe malaria (SM) is a life-threatening complication of infection with Plasmodium falciparum Epidemiological observations have long indicated that immunity against SM is acquired relatively rapidly, but prospective studies to investigate its immunological basis are logistically challenging and have rarely been undertaken. We investigated the merozoite targets and antibody-mediated mechanisms associated with protection against SM in Kenyan children aged 0 to 2 years. We designed a unique prospective matched case-control study of well-characterized SM clinical phenotypes nested within a longitudinal birth cohort of children (n= 5,949) monitored over the first 2 years of life. We quantified immunological parameters in sera collected before the SM event in cases and their individually matched controls to evaluate the prospective odds of developing SM in the first 2 years of life. Anti-AMA1 antibodies were associated with a significant reduction in the odds of developing SM (odds ratio [OR] = 0.37; 95% confidence interval [CI] = 0.15 to 0.90; P= 0.029) after adjustment for responses to all other merozoite antigens tested, while those against MSP-2, MSP-3, Plasmodium falciparum Rh2 [PfRh2], MSP-119, and the infected red blood cell surface antigens were not. The combined ability of total IgG to inhibit parasite growth and mediate the release of reactive oxygen species from neutrophils was associated with a marked reduction in the odds of developing SM (OR = 0.07; 95% CI = 0.006 to 0.82;P= 0.03). Assays of these two functional mechanisms were poorly correlated (Spearman rank correlation coefficient [rs] = 0.12;P= 0.07). Our data provide epidemiological evidence that multiple antibody-dependent mechanisms contribute to protective immunity via distinct targets whose identification could accelerate the development of vaccines to protect against SM.


Subject(s)
Antibodies, Protozoan/physiology , Malaria, Falciparum/immunology , Plasmodium falciparum/physiology , Aging , Antigens, Protozoan/immunology , Case-Control Studies , Child , Child, Preschool , Growth Inhibitors/metabolism , Humans , Infant , Kenya/epidemiology , Merozoites/immunology , Odds Ratio , Respiratory Burst/physiology
12.
Vaccine ; 34(1): 160-6, 2016 Jan 02.
Article in English | MEDLINE | ID: mdl-26541134

ABSTRACT

BACKGROUND: Young infants have reduced susceptibility to febrile malaria compared with older children, but the mechanism for this remains unclear. There are conflicting data on the role of passively acquired antibodies. Here, we examine antibody titres to merozoite surface antigens in the protection of children in their first two years of life in two settings with differing malaria transmission intensity and compare these titres to previously established protective thresholds. METHODS: Two cohorts of children aged four to six weeks were recruited in Banfora, Burkina and Keur Soce, Senegal and followed up for two years. Malaria infections were detected by light microscopic examination of blood smears collected at active and passive case detection visits. The titres of antibodies to the Plasmodium falciparum recombinant merozoite proteins (AMA1-3D7, MSP1-19, MSP2-Dd2, and MSP3-3D7) were measured by enzyme-linked immunosorbent assay at 1-6, 9, 12, 15 and 18 months of age and compared with the protective thresholds established in Kenyan children. RESULTS: Antibody titres were below the protective thresholds throughout the study period and we did not find any association with protection against febrile malaria. Antibodies to AMA1 and MSP1-19 appeared to be markers of exposure in the univariate analysis (and so associated with increasing risk) and adjusting for exposure reduced the strength and significance of this association. CONCLUSION: The antibody levels we measured are unlikely to be responsible for the apparent protection against febrile malaria seen in young infants. Further work to identify protective antibody responses might include functional assays and a wider range of antigens.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria/immunology , Merozoites/immunology , Plasmodium falciparum/immunology , Burkina Faso/epidemiology , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Longitudinal Studies , Malaria/epidemiology , Malaria/transmission , Male , Prospective Studies , Senegal/epidemiology
13.
Sci Rep ; 5: 14081, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26373337

ABSTRACT

The assessment of naturally-acquired and vaccine-induced immunity to blood-stage Plasmodium falciparum malaria is of long-standing interest. However, the field has suffered from a paucity of in vitro assays that reproducibly measure the anti-parasitic activity induced by antibodies in conjunction with immune cells. Here we optimize the antibody-dependent respiratory burst (ADRB) assay, which assesses the ability of antibodies to activate the release of reactive oxygen species from human neutrophils in response to P. falciparum blood-stage parasites. We focus particularly on assay parameters affecting serum preparation and concentration, and importantly assess reproducibility. Our standardized protocol involves testing each serum sample in singlicate with three independent neutrophil donors, and indexing responses against a standard positive control of pooled hyper-immune Kenyan sera. The protocol can be used to quickly screen large cohorts of samples from individuals enrolled in immuno-epidemiological studies or clinical vaccine trials, and requires only 6 µL of serum per sample. Using a cohort of 86 samples, we show that malaria-exposed individuals induce higher ADRB activity than malaria-naïve individuals. The development of the ADRB assay complements the use of cell-independent assays in blood-stage malaria, such as the assay of growth inhibitory activity, and provides an important standardized cell-based assay in the field.


Subject(s)
Antibodies, Protozoan/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Neutrophils/immunology , Plasmodium falciparum/immunology , Respiratory Burst/immunology , Adult , Humans , In Vitro Techniques , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Neutrophils/metabolism , Receptors, Fc/metabolism , Reproducibility of Results
14.
BMC Med ; 13: 114, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25967134

ABSTRACT

BACKGROUND: Epidemiological studies indicate that some children experience many more episodes of clinical malaria than their age mates in a given location. Whether this is as a result of the micro-heterogeneity of malaria transmission with some children effectively getting more exposure to infectious mosquitoes than others, or reflects a failure in the acquisition of immunity needs to be elucidated. Here, we investigated the determinants of increased susceptibility to clinical malaria by comparing the intensity of exposure to Plasmodium falciparum and the acquisition of immunity in children at the extreme ends of the over-dispersed distribution of the incidence of clinical malaria. METHODS: The study was nested within a larger cohort in an area where the intensity of malaria transmission was low. We identified children who over a five-year period experienced 5 to 16 clinical malaria episodes (children at the tail-end of the over-dispersed distribution, n = 35), remained malaria-free (n = 12) or had a single episode (n = 26). We quantified antibodies against seven Plasmodium falciparum merozoite antigens in plasma obtained at six cross-sectional surveys spanning these five years. We analyzed the antibody responses to identify temporal dynamics that associate with disease susceptibility. RESULTS: Children experiencing multiple episodes of malaria were more likely to be parasite positive by microscopy at cross-sectional surveys (X (2) test for trend 14.72 P = 0.001) and had a significantly higher malaria exposure index, than those in the malaria-free or single episode groups (Kruskal-Wallis test P = 0.009). In contrast, the five-year temporal dynamics of anti-merozoite antibodies were similar in the three groups. Importantly in all groups, antibody levels were below the threshold concentrations previously observed to be correlated with protective immunity. CONCLUSIONS: We conclude that in the context of a low malaria transmission setting, susceptibility to clinical malaria is not accounted for by anti-merozoite antibodies but appears to be a consequence of increased parasite exposure. We hypothesize that intensive exposure is a prerequisite for protective antibody concentrations, while little to modest exposure may manifest as multiple clinical infections with low levels of antibodies. These findings have implications for interventions that effectively lower malaria transmission intensity.


Subject(s)
Malaria, Falciparum/epidemiology , Malaria, Falciparum/immunology , Animals , Antibodies, Protozoan/blood , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Incidence , Infant , Malaria, Falciparum/transmission , Male , Plasmodium falciparum
15.
Vaccine ; 31(37): 3936-42, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23800539

ABSTRACT

Antibodies to selected Plasmodium falciparum merozoite antigens are often reported to be associated with protection from malaria in one epidemiological cohort, but not in another. Here, we sought to understand this paradox by exploring the hypothesis that a threshold concentration of antibodies is necessary for protection. We analyzed data from two independent cohorts along the Kenyan coast, one in which antibodies to AMA1, MSP-2 and MSP-3 were associated with protection from malaria (Chonyi) and another in which this association was not observed (Junju). We used a malaria reference reagent to standardize antibody measurements across both cohorts, and applied statistical methods to derive the threshold concentration of antibodies against each antigen that best correlated with a reduced risk of malaria (the protective threshold), in the Chonyi cohort. We then tested whether antibodies in Junju reached the protective threshold concentrations observed in the Chonyi cohort. Except for children under 3 years, the age-matched proportions of children achieving protective threshold concentrations of antibodies against AMA1 and MSP-2 were significantly lower in Junju compared to Chonyi (Fishers exact test, P<0.01). For MSP-3, this difference was significant only among 4-5 year olds. We conclude that although antibodies are commonly detected in malaria endemic populations, they may be present in concentrations that are insufficient for protection. Our results have implications for the analysis and interpretation of similar data from immuno-epidemiological studies.


Subject(s)
Antibodies, Protozoan/immunology , Malaria/immunology , Malaria/prevention & control , Merozoites/immunology , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Child , Child, Preschool , Cohort Studies , Humans , Infant , Infant, Newborn , Kenya , Malaria/parasitology , Malaria/transmission , Plasmodium falciparum/immunology , Risk Factors
16.
PLoS One ; 8(2): e57320, 2013.
Article in English | MEDLINE | ID: mdl-23437368

ABSTRACT

BACKGROUND: Plasmodium falciparum malaria remains a major cause of illness and death in sub-Saharan Africa. Young children bear the brunt of the disease and though older children and adults suffer relatively fewer clinical attacks, they remain susceptible to asymptomatic P. falciparum infection. A better understanding of the host factors associated with immunity to clinical malaria and the ability to sustain asymptomatic P. falciparum infection will aid the development of improved strategies for disease prevention. METHODS AND FINDINGS: Here we investigate whether full differential blood counts can predict susceptibility to clinical malaria among Kenyan children sampled at five annual cross-sectional surveys. We find that the ratio of monocytes to lymphocytes, measured in peripheral blood at the time of survey, directly correlates with risk of clinical malaria during follow-up. This association is evident among children with asymptomatic P. falciparum infection at the time the cell counts are measured (Hazard ratio (HR)  =  2.7 (95% CI 1.42, 5.01, P  =  0.002) but not in those without detectable parasitaemia (HR  =  1.0 (95% CI 0.74, 1.42, P  =  0.9). CONCLUSIONS: We propose that the monocyte to lymphocyte ratio, which is easily derived from routine full differential blood counts, reflects an individual's capacity to mount an effective immune response to P. falciparum infection.


Subject(s)
Lymphocytes/immunology , Malaria, Falciparum/immunology , Monocytes/immunology , Parasitemia/immunology , Plasmodium falciparum/immunology , Asymptomatic Infections , Blood Cell Count , Child , Child, Preschool , Cross-Sectional Studies , Disease Susceptibility , Female , Genotype , Humans , Kenya , Lymphocytes/pathology , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Male , Monocytes/pathology , Parasitemia/diagnosis , Parasitemia/parasitology , Parasitemia/pathology , Prognosis , Risk
17.
PLoS One ; 8(2): e56032, 2013.
Article in English | MEDLINE | ID: mdl-23418502

ABSTRACT

Children in malaria endemic areas acquire immunity to severe malaria faster than to mild malaria. Only a minority of children suffers from severe malaria and it is not known what determines this. The aim of this study was to establish how P. falciparum infections during the first years of life affect the risk of severe malaria. A matched case-control study was nested within a large birth cohort set up to study the immunoepidemiology of pneumococci on the Kenyan coast. Infection patterns in three-monthly blood samples in cohort children admitted to hospital with severe malaria were compared to controls matched on age, residential location and time of sampling. P. falciparum detected at least once from birth conferred an increased risk of severe malaria and particularly if multiclonal infections, as characterized by genotyping of a polymorphic antigen gene, were ever detected. The results show for the first time that children with severe malaria have more infections early in life compared to community controls. These findings provide important insights on the immunity to severe disease, knowledge essential for the development of a vaccine against severe malaria.


Subject(s)
Malaria, Falciparum/epidemiology , Plasmodium falciparum/isolation & purification , Case-Control Studies , Child, Preschool , Female , Humans , Infant , Kenya/epidemiology , Malaria, Falciparum/immunology , Male , Risk , Severity of Illness Index
18.
PLoS Pathog ; 8(11): e1002991, 2012.
Article in English | MEDLINE | ID: mdl-23144611

ABSTRACT

No vaccine has yet proven effective against the blood-stages of Plasmodium falciparum, which cause the symptoms and severe manifestations of malaria. We recently found that PfRH5, a P. falciparum-specific protein expressed in merozoites, is efficiently targeted by broadly-neutralizing, vaccine-induced antibodies. Here we show that antibodies against PfRH5 efficiently inhibit the in vitro growth of short-term-adapted parasite isolates from Cambodia, and that the EC(50) values of antigen-specific antibodies against PfRH5 are lower than those against PfAMA1. Since antibody responses elicited by multiple antigens are speculated to improve the efficacy of blood-stage vaccines, we conducted detailed assessments of parasite growth inhibition by antibodies against PfRH5 in combination with antibodies against seven other merozoite antigens. We found that antibodies against PfRH5 act synergistically with antibodies against certain other merozoite antigens, most notably with antibodies against other erythrocyte-binding antigens such as PfRH4, to inhibit the growth of a homologous P. falciparum clone. A combination of antibodies against PfRH4 and basigin, the erythrocyte receptor for PfRH5, also potently inhibited parasite growth. This methodology provides the first quantitative evidence that polyclonal vaccine-induced antibodies can act synergistically against P. falciparum antigens and should help to guide the rational development of future multi-antigen vaccines.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Carrier Proteins/immunology , Erythrocytes/immunology , Merozoites/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Erythrocytes/parasitology , Humans , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Mice
19.
J Immunol ; 187(6): 2885-97, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21880980

ABSTRACT

IFN-γ and T cells are both required for the development of experimental cerebral malaria during Plasmodium berghei ANKA infection. Surprisingly, however, the role of IFN-γ in shaping the effector CD4(+) and CD8(+) T cell response during this infection has not been examined in detail. To address this, we have compared the effector T cell responses in wild-type and IFN-γ(-/-) mice during P. berghei ANKA infection. The expansion of splenic CD4(+) and CD8(+) T cells during P. berghei ANKA infection was unaffected by the absence of IFN-γ, but the contraction phase of the T cell response was significantly attenuated. Splenic T cell activation and effector function were essentially normal in IFN-γ(-/-) mice; however, the migration to, and accumulation of, effector CD4(+) and CD8(+) T cells in the lung, liver, and brain was altered in IFN-γ(-/-) mice. Interestingly, activation and accumulation of T cells in various nonlymphoid organs was differently affected by lack of IFN-γ, suggesting that IFN-γ influences T cell effector function to varying levels in different anatomical locations. Importantly, control of splenic T cell numbers during P. berghei ANKA infection depended on active IFN-γ-dependent environmental signals--leading to T cell apoptosis--rather than upon intrinsic alterations in T cell programming. To our knowledge, this is the first study to fully investigate the role of IFN-γ in modulating T cell function during P. berghei ANKA infection and reveals that IFN-γ is required for efficient contraction of the pool of activated T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Malaria/immunology , Plasmodium berghei/immunology , Animals , Cell Movement/immunology , Cell Separation , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
20.
Infect Immun ; 78(11): 4625-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20732997

ABSTRACT

Although Plasmodium falciparum apical membrane antigen 1 (AMA1) is a leading malaria vaccine candidate, extensive allelic diversity may compromise its vaccine potential. We have previously shown that naturally acquired antibodies to AMA1 were associated with protection from clinical malaria in this Kenyan population. To assess the impact of allelic diversity on naturally acquired immunity, we first sequenced the ectodomain-encoding region of P. falciparum ama1 from subjects with asymptomatic, mild, and severe malaria and measured allele frequency distributions. We then measured antibodies to three allelic AMA1 proteins (AMA1_3D7, AMA1_FVO, and AMA1_HB3) and used competition enzyme-linked immunosorbent assays (ELISAs) to analyze allele-specific antibodies. Seventy-eight unique haplotypes were identified from 129 alleles sampled. No clustering of allelic haplotypes with disease severity or year of sampling was observed. Differences in nucleotide frequencies in clinical (severe plus mild malaria) versus asymptomatic infections were observed at 16 polymorphic positions. Allele frequency distributions were indicative of balancing selection, with the strongest signature being identified in domain III (Tajima's D = 2.51; P < 0.05). Antibody reactivities to each of the three allelic AMA1 proteins were highly correlated (P < 0.001 for all pairwise comparisons). Although antibodies to conserved epitopes were abundant, 48% of selected children with anti-AMA1 IgG (n = 106) had detectable reactivity to allele-specific epitopes as determined by a competition ELISA. Antibodies to both conserved and allele-specific epitopes in AMA1 may contribute to clinical protection.


Subject(s)
Alleles , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Genetic Variation , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Membrane Proteins/immunology , Protozoan Proteins/immunology , Adaptive Immunity , Amino Acid Sequence , Case-Control Studies , Child , Child, Preschool , Gene Frequency , Haplotypes , Humans , Infant , Kenya , Malaria, Falciparum/parasitology , Malaria, Falciparum/physiopathology , Molecular Sequence Data , Plasmodium falciparum/immunology , Sequence Analysis, DNA , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...