Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Clin Infect Dis ; 64(5): 645-653, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28362910

ABSTRACT

Background: Chronic asymptomatic Plasmodium falciparum infections are common in endemic areas and are thought to contribute to the maintenance of malaria immunity. Whether treatment of these infections increases the subsequent risk of clinical episodes of malaria is unclear. Methods: In a 3-year study in Mali, asymptomatic individuals with or without P. falciparum infection at the end of the 6-month dry season were identified by polymerase chain reaction (PCR), and clinical malaria risk was compared during the ensuing 6-month malaria transmission season. At the end of the second dry season, 3 groups of asymptomatic children were identified: (1) children infected with P. falciparum as detected by rapid diagnostic testing (RDT) who were treated with antimalarials (n = 104), (2) RDT-negative children whose untreated P. falciparum infections were detected retrospectively by PCR (n = 55), and (3) uninfected children (RDT/PCR negative) (n = 434). Clinical malaria risk during 2 subsequent malaria seasons was compared. Plasmodium falciparum-specific antibody kinetics during the dry season were compared in children who did or did not harbor asymptomatic P. falciparum infections. Results: Chronic asymptomatic P. falciparum infection predicted decreased clinical malaria risk during the subsequent malaria season(s); treatment of these infections did not alter this reduced risk. Plasmodium falciparum-specific antibodies declined similarly in children who did or did not harbor chronic asymptomatic P. falciparum infection during the dry season. Conclusions: These findings challenge the notion that chronic asymptomatic P. falciparum infection maintains malaria immunity and suggest that mass drug administration during the dry season should not increase the subsequent risk of clinical malaria.


Subject(s)
Malaria, Falciparum/epidemiology , Plasmodium falciparum , Adolescent , Adult , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antimalarials/pharmacology , Antimalarials/therapeutic use , Asymptomatic Infections , Child , Child, Preschool , Chronic Disease , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Infant , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Male , Mali/epidemiology , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Population Surveillance , Risk , Seasons , Young Adult
2.
Elife ; 42015 May 08.
Article in English | MEDLINE | ID: mdl-25955968

ABSTRACT

Protective antibodies in Plasmodium falciparum malaria are only acquired after years of repeated infections. Chronic malaria exposure is associated with a large increase in atypical memory B cells (MBCs) that resemble B cells expanded in a variety of persistent viral infections. Understanding the function of atypical MBCs and their relationship to classical MBCs will be critical to developing effective vaccines for malaria and other chronic infections. We show that VH gene repertoires and somatic hypermutation rates of atypical and classical MBCs are indistinguishable indicating a common developmental history. Atypical MBCs express an array of inhibitory receptors and B cell receptor (BCR) signaling is stunted in atypical MBCs resulting in impaired B cell responses including proliferation, cytokine production and antibody secretion. Thus, in response to chronic malaria exposure, atypical MBCs appear to differentiate from classical MBCs becoming refractory to BCR-mediated activation and potentially interfering with the acquisition of malaria immunity.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Malaria, Falciparum/immunology , Malaria/immunology , Signal Transduction/immunology , Adolescent , Adult , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , B-Lymphocytes/parasitology , B-Lymphocytes/pathology , Child , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/immunology , Female , Gene Expression , Host-Parasite Interactions , Humans , Immunoglobulin E/biosynthesis , Immunoglobulin M/biosynthesis , Immunophenotyping , Malaria/parasitology , Malaria/pathology , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Male , Middle Aged , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology
3.
PLoS Negl Trop Dis ; 8(9): e3154, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25210876

ABSTRACT

BACKGROUND: Malaria and schistosomiasis often overlap in tropical and subtropical countries and impose tremendous disease burdens; however, the extent to which schistosomiasis modifies the risk of febrile malaria remains unclear. METHODS: We evaluated the effect of baseline S. haematobium mono-infection, baseline P. falciparum mono-infection, and co-infection with both parasites on the risk of febrile malaria in a prospective cohort study of 616 children and adults living in Kalifabougou, Mali. Individuals with S. haematobium were treated with praziquantel within 6 weeks of enrollment. Malaria episodes were detected by weekly physical examination and self-referral for 7 months. The primary outcome was time to first or only malaria episode defined as fever (≥ 37.5 °C) and parasitemia (≥ 2500 asexual parasites/µl). Secondary definitions of malaria using different parasite densities were also explored. RESULTS: After adjusting for age, anemia status, sickle cell trait, distance from home to river, residence within a cluster of high S. haematobium transmission, and housing type, baseline P. falciparum mono-infection (n = 254) and co-infection (n = 39) were significantly associated with protection from febrile malaria by Cox regression (hazard ratios 0.71 and 0.44; P = 0.01 and 0.02; reference group: uninfected at baseline). Baseline S. haematobium mono-infection (n = 23) did not associate with malaria protection in the adjusted analysis, but this may be due to lack of statistical power. Anemia significantly interacted with co-infection (P = 0.009), and the malaria-protective effect of co-infection was strongest in non-anemic individuals. Co-infection was an independent negative predictor of lower parasite density at the first febrile malaria episode. CONCLUSIONS: Co-infection with S. haematobium and P. falciparum is significantly associated with reduced risk of febrile malaria in long-term asymptomatic carriers of P. falciparum. Future studies are needed to determine whether co-infection induces immunomodulatory mechanisms that protect against febrile malaria or whether genetic, behavioral, or environmental factors not accounted for here explain these findings.


Subject(s)
Coinfection/parasitology , Malaria, Falciparum/parasitology , Plasmodium falciparum/isolation & purification , Schistosoma haematobium/isolation & purification , Schistosomiasis haematobia/parasitology , Adolescent , Adult , Animals , Child , Child, Preschool , Coinfection/epidemiology , Female , Humans , Infant , Malaria, Falciparum/epidemiology , Male , Mali/epidemiology , Prospective Studies , Schistosomiasis haematobia/epidemiology , Young Adult
4.
PLoS Pathog ; 10(4): e1004079, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24743880

ABSTRACT

In malaria-naïve individuals, Plasmodium falciparum infection results in high levels of parasite-infected red blood cells (iRBCs) that trigger systemic inflammation and fever. Conversely, individuals in endemic areas who are repeatedly infected are often asymptomatic and have low levels of iRBCs, even young children. We hypothesized that febrile malaria alters the immune system such that P. falciparum re-exposure results in reduced production of pro-inflammatory cytokines/chemokines and enhanced anti-parasite effector responses compared to responses induced before malaria. To test this hypothesis we used a systems biology approach to analyze PBMCs sampled from healthy children before the six-month malaria season and the same children seven days after treatment of their first febrile malaria episode of the ensuing season. PBMCs were stimulated with iRBC in vitro and various immune parameters were measured. Before the malaria season, children's immune cells responded to iRBCs by producing pro-inflammatory mediators such as IL-1ß, IL-6 and IL-8. Following malaria there was a marked shift in the response to iRBCs with the same children's immune cells producing lower levels of pro-inflammatory cytokines and higher levels of anti-inflammatory cytokines (IL-10, TGF-ß). In addition, molecules involved in phagocytosis and activation of adaptive immunity were upregulated after malaria as compared to before. This shift was accompanied by an increase in P. falciparum-specific CD4+Foxp3- T cells that co-produce IL-10, IFN-γ and TNF; however, after the subsequent six-month dry season, a period of markedly reduced malaria transmission, P. falciparum-inducible IL-10 production remained partially upregulated only in children with persistent asymptomatic infections. These findings suggest that in the face of P. falciparum re-exposure, children acquire exposure-dependent P. falciparum-specific immunoregulatory responses that dampen pathogenic inflammation while enhancing anti-parasite effector mechanisms. These data provide mechanistic insight into the observation that P. falciparum-infected children in endemic areas are often afebrile and tend to control parasite replication.


Subject(s)
Adaptive Immunity , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adolescent , CD4-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Cytokines/blood , Female , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/epidemiology , Male , Plasmodium falciparum/metabolism
5.
J Infect Dis ; 209(5): 789-98, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24133188

ABSTRACT

BACKGROUND: Plasmodium falciparum reticulocyte-binding protein homologue 5 (PfRH5) is a blood-stage parasite protein essential for host erythrocyte invasion. PfRH5-specific antibodies raised in animals inhibit parasite growth in vitro, but the relevance of naturally acquired PfRH5-specific antibodies in humans is unclear. METHODS: We assessed pre-malaria season PfRH5-specific immunoglobulin G (IgG) levels in 357 Malian children and adults who were uninfected with Plasmodium. Subsequent P. falciparum infections were detected by polymerase chain reaction every 2 weeks and malaria episodes by weekly physical examination and self-referral for 7 months. The primary outcome was time between the first P. falciparum infection and the first febrile malaria episode. PfRH5-specific IgG was assayed for parasite growth-inhibitory activity. RESULTS: The presence of PfRH5-specific IgG at enrollment was associated with a longer time between the first blood-stage infection and the first malaria episode (PfRH5-seropositive median: 71 days, PfRH5-seronegative median: 18 days; P = .001). This association remained significant after adjustment for age and other factors associated with malaria risk/exposure (hazard ratio, .62; P = .02). Concentrated PfRH5-specific IgG purified from Malians inhibited P. falciparum growth in vitro. CONCLUSIONS: Naturally acquired PfRH5-specific IgG inhibits parasite growth in vitro and predicts protection from malaria. These findings strongly support efforts to develop PfRH5 as an urgently needed blood-stage malaria vaccine. CLINICAL TRIALS REGISTRATION: NCT01322581.


Subject(s)
Antibodies, Protozoan/immunology , Carrier Proteins/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Immunoglobulin G/immunology , Infant , Malaria Vaccines/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Male , Placenta/immunology , Placenta/parasitology , Pregnancy , Reticulocytes/immunology , Reticulocytes/parasitology , Young Adult
6.
Clin Infect Dis ; 57(1): 40-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23487390

ABSTRACT

BACKGROUND: In experimental models of human and mouse malaria, sterilizing liver stage immunity that blocks progression of Plasmodium infection to the symptomatic blood stage can be readily demonstrated. However, it remains unclear whether individuals in malaria-endemic areas acquire such immunity. METHODS: In Mali, 251 healthy children and adults aged 4-25 years who were free of blood-stage Plasmodium infection by polymerase chain reaction (PCR) were enrolled in a longitudinal study just prior to an intense 6-month malaria season. Subsequent clinical malaria episodes were detected by weekly active surveillance and self-referral. Asymptomatic P. falciparum infections were detected by blood-smear microscopy and PCR analysis of dried blood spots that had been collected every 2 weeks for 7 months. RESULTS: As expected, the risk of clinical malaria decreased with increasing age (log-rank test, P = .0038). However, analysis of PCR data showed no age-related differences in P. falciparum infection risk (log-rank test, P = .37). CONCLUSIONS: Despite years of exposure to intense P. falciparum transmission, there is no evidence of acquired, sterile immunity to P. falciparum infection in this population, even as clinical immunity to blood-stage malaria is clearly acquired. Understanding why repeated P. falciparum infections do not induce sterile protection may lead to insights for developing vaccines that target the liver stage in malaria-endemic populations.


Subject(s)
Adaptive Immunity , Malaria, Falciparum/epidemiology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adult , Child , Cohort Studies , Humans , Longitudinal Studies , Malaria, Falciparum/prevention & control , Mali/epidemiology
7.
PLoS Pathog ; 6(5): e1000912, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20502681

ABSTRACT

Immunity to Plasmodium falciparum (Pf) malaria is only acquired after years of repeated infections and wanes rapidly without ongoing parasite exposure. Antibodies are central to malaria immunity, yet little is known about the B-cell biology that underlies the inefficient acquisition of Pf-specific humoral immunity. This year-long prospective study in Mali of 185 individuals aged 2 to 25 years shows that Pf-specific memory B-cells and antibodies are acquired gradually in a stepwise fashion over years of repeated Pf exposure. Both Pf-specific memory B cells and antibody titers increased after acute malaria and then, after six months of decreased Pf exposure, contracted to a point slightly higher than pre-infection levels. This inefficient, stepwise expansion of both the Pf-specific memory B-cell and long-lived antibody compartments depends on Pf exposure rather than age, based on the comparator response to tetanus vaccination that was efficient and stable. These observations lend new insights into the cellular basis of the delayed acquisition of malaria immunity.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Immunologic Memory/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Acute Disease , Adolescent , Adult , Antibodies, Protozoan/blood , Child , Child, Preschool , Female , Humans , Immunoglobulin G/blood , Immunophenotyping , Longitudinal Studies , Malaria, Falciparum/transmission , Male , Mali , Prospective Studies , Recurrence , Seasons , Young Adult
8.
Proc Natl Acad Sci U S A ; 107(15): 6958-63, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20351286

ABSTRACT

Abs are central to malaria immunity, which is only acquired after years of exposure to Plasmodium falciparum (Pf). Despite the enormous worldwide burden of malaria, the targets of protective Abs and the basis of their inefficient acquisition are unknown. Addressing these knowledge gaps could accelerate malaria vaccine development. To this end, we developed a protein microarray containing approximately 23% of the Pf 5,400-protein proteome and used this array to probe plasma from 220 individuals between the ages of 2-10 years and 18-25 years in Mali before and after the 6-month malaria season. Episodes of malaria were detected by passive surveillance over the 8-month study period. Ab reactivity to Pf proteins rose dramatically in children during the malaria season; however, most of this response appeared to be short-lived based on cross-sectional analysis before the malaria season, which revealed only modest incremental increases in Ab reactivity with age. Ab reactivities to 49 Pf proteins measured before the malaria season were significantly higher in 8-10-year-old children who were infected with Pf during the malaria season but did not experience malaria (n = 12) vs. those who experienced malaria (n = 29). This analysis also provided insight into patterns of Ab reactivity against Pf proteins based on the life cycle stage at which proteins are expressed, subcellular location, and other proteomic features. This approach, if validated in larger studies and in other epidemiological settings, could prove to be a useful strategy for better understanding fundamental properties of the human immune response to Pf and for identifying previously undescribed vaccine targets.


Subject(s)
Malaria, Falciparum/immunology , Plasmodium falciparum/metabolism , Protein Array Analysis/methods , Adolescent , Adult , Animals , Antigens, Protozoan/immunology , Child , Child, Preschool , Cohort Studies , Humans , Immune System , Malaria Vaccines/chemistry , Mali , Proteomics/methods
9.
Infect Immun ; 78(2): 737-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19917712

ABSTRACT

Immunity to the asexual blood stage of Plasmodium falciparum is complex and likely involves several effector mechanisms. Antibodies are thought to play a critical role in malaria immunity, and a corresponding in vitro correlate of antibody-mediated immunity has long been sought to facilitate malaria vaccine development. The growth inhibition assay (GIA) measures the capacity of antibodies to limit red blood cell (RBC) invasion and/or growth of P. falciparum in vitro. In humans, naturally acquired and vaccine-induced P. falciparum-specific antibodies have growth-inhibitory activity, but it is unclear if growth-inhibitory activity correlates with protection from clinical disease. In a longitudinal study in Mali, purified IgGs, obtained from plasmas collected before the malaria season from 220 individuals aged 2 to 10 and 18 to 25 years, were assayed for growth-inhibitory activity. Malaria episodes were recorded by passive surveillance over the subsequent 6-month malaria season. Logistic regression showed that greater age (odds ratio [OR], 0.78; 95% confidence interval [95% CI], 0.63 to 0.95; P = 0.02) and growth-inhibitory activity (OR, 0.50; 95% CI, 0.30 to 0.85; P = 0.01) were significantly associated with decreased malaria risk in children. A growth-inhibitory activity level of 40% was determined to be the optimal cutoff for discriminating malaria-immune and susceptible individuals in this cohort, with a sensitivity of 97.0%, but a low specificity of 24.3%, which limited the assay's ability to accurately predict protective immunity and to serve as an in vitro correlate of antibody-mediated immunity. These data suggest that antibodies which block merozoite invasion of RBC and/or inhibit the intra-RBC growth of the parasite contribute to but are not sufficient for the acquisition of malaria immunity.


Subject(s)
Antibodies, Protozoan/immunology , Malaria/immunology , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Female , Hematologic Tests/methods , Humans , Immunoglobulin G/immunology , In Vitro Techniques , Male , Mali , Plasmodium malariae/immunology , Risk , Young Adult
10.
Vaccine ; 27(52): 7299-303, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-19712767

ABSTRACT

Antibodies play a key role in controlling blood stage malaria infections, and an effective blood stage malaria vaccine will likely require that it induce vaccine-specific memory B cells (MBCs). Our previous studies showed that the addition of the TLR9 agonist CpG to Plasmodium falciparum protein subunit vaccines greatly increased their efficacy in inducing MBCs in nonimmune U.S. volunteers. Here we show that in contrast the same CpG-containing malaria vaccine did not enhance the acquisition of MBCs in semi-immune adults living in Mali. Understanding the molecular basis of this apparent refractoriness to TLR9 agonist will be of significant interest in vaccine design.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 9/antagonists & inhibitors , Adjuvants, Immunologic/pharmacology , Adult , Antibodies, Protozoan/blood , Flow Cytometry , Humans , Malaria, Falciparum/immunology , Mali , Oligodeoxyribonucleotides/immunology , Plasmodium falciparum/immunology , Time Factors , Vaccines, Subunit/immunology
11.
Malar J ; 8: 5, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19128455

ABSTRACT

BACKGROUND: Because of the emergence of chloroquine resistance in Mali, artemether-lumefantrine (AL) or artesunate-amodiaquine (AS+AQ) are recommended as first-line therapy for uncomplicated malaria, but have not been available in Mali until recently because of high costs. METHODS: From July 2005 to January 2006, a randomized open-label trial of three oral antimalarial combinations, namely AS+AQ, artesunate plus sulphadoxine-pyrimethamine (AS+SP), and amodiaquine plus sulphadoxine-pyrimethamine (AQ+SP), was conducted in Faladje, Mali. Parasite genotyping by polymerase chain reaction (PCR) was used to distinguish new from recrudescent Plasmodium falciparum infections. RESULTS: 397 children 6 to 59 months of age with uncomplicated Plasmodium falciparum malaria were enrolled, and followed for 28 days to assess treatment efficacy. Baseline characteristics were similar in all three treatment groups. The uncorrected rates of adequate clinical and parasitologic response (ACPR) were 55.7%, 90.8%, and 97.7% in AS+AQ, AS+SP, and AQ+SP respectively (p < 0.001); after PCR correction ACPR rates were similar among treatment groups: 95.4%, 96.9%, and 99.2% respectively (p = 0.17). Mean haemoglobin concentration increased across all treatment groups from Day 0 (9.82 +/- 1.68 g/dL) to Day 28 (10.78 +/- 1.49 g/dL) (p < 0.001), with the greatest improvement occurring in children treated with AQ+SP. On Day 2, the prevalence of parasitaemia was significantly greater among children treated with AQ+SP (50.8%) than in children treated with AS+AQ (10.5%) or AS+SP (10.8%) (p < 0.001). No significant difference in gametocyte carriage was found between groups during the follow-up period. CONCLUSION: The combination of AQ+SP provides a potentially low cost alternative for treatment of uncomplicated P. falciparum infection in Mali and appears to have the added value of longer protective effect against new infection.


Subject(s)
Amodiaquine/therapeutic use , Antimalarials/therapeutic use , Artemisinins/therapeutic use , Malaria, Falciparum/drug therapy , Pyrimethamine/therapeutic use , Sulfadoxine/therapeutic use , Amodiaquine/administration & dosage , Amodiaquine/adverse effects , Animals , Antimalarials/administration & dosage , Antimalarials/adverse effects , Artemisinins/administration & dosage , Artemisinins/adverse effects , Child, Preschool , Drug Combinations , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Infant , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Male , Mali/epidemiology , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Polymerase Chain Reaction , Pyrimethamine/administration & dosage , Pyrimethamine/adverse effects , Single-Blind Method , Sulfadoxine/administration & dosage , Sulfadoxine/adverse effects , Treatment Outcome
12.
J Infect Dis ; 198(9): 1265-75, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18752444

ABSTRACT

BACKGROUND: The World Health Organization (WHO) recently recommended that the time to first malaria episode serve as the primary end point in phase III malaria vaccine trials--the first of which will be held in Africa. Although common red blood cell (RBC) polymorphisms such as sickle hemoglobin (HbS) are known to protect against malaria in Africa, their impact on this end point has not been investigated. METHODS: A longitudinal study of 225 individuals aged 2-25 years was conducted in Mali. The association between common RBC polymorphisms and the time to first malaria episode was evaluated. RESULTS: Among children aged 2-10 years, sickle cell trait (HbAS) was associated with a 34-day delay in the median time to first malaria episode (P= .017) Cox regression analysis showed that greater age (hazard ratio [HR], 0.87 [95% CI, 0.80-0.94]; (P= .001), HbAS (HR, 0.48 [95% CI, 0.26-0.91]; (P= .024), and asymptomatic parasitemia at enrollment (HR, 0.35 [95% CI, 0.14-0.85]; (P= .021) were associated with decreased malaria risk. CONCLUSION: Given the delay in the time to first malaria episode associated with HbAS, it would be advisable for clinical trials and observational studies that use this end point to include Hb typing in the design of studies conducted in areas where HbAS is prevalent.


Subject(s)
Malaria, Falciparum/genetics , Sickle Cell Trait/genetics , Adolescent , Adult , Blood Group Antigens , Child , Child, Preschool , Clinical Trials as Topic , Cohort Studies , Erythrocytes , Female , Hemoglobins/genetics , Humans , Male , Multivariate Analysis , Parasitemia , Polymorphism, Genetic , Prevalence , Risk Factors , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...