Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 32: 157-87, 2014.
Article in English | MEDLINE | ID: mdl-24655294

ABSTRACT

Malaria is a mosquito-borne disease caused by parasites of the obligate intracellular Apicomplexa phylum the most deadly of which, Plasmodium falciparum, prevails in Africa. Malaria imposes a huge health burden on the world's most vulnerable populations, claiming the lives of nearly one million children and pregnant women each year. Although there is keen interest in eradicating malaria, we do not yet have the necessary tools to meet this challenge, including an effective malaria vaccine and adequate vector control strategies. Here we review what is known about the mechanisms at play in immune resistance to malaria in both the human and mosquito hosts at each step in the parasite's complex life cycle with a view toward developing the tools that will contribute to the prevention of disease and death and, ultimately, to the goal of malaria eradication. In so doing, we hope to inspire immunologists to participate in defeating this devastating disease.


Subject(s)
Culicidae/immunology , Host-Pathogen Interactions/immunology , Malaria/immunology , Plasmodium/immunology , Animals , Culicidae/parasitology , Humans , Life Cycle Stages , Malaria/parasitology , Malaria/prevention & control , Plasmodium/growth & development , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology
2.
Cell ; 159(6): 1277-89, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25480293

ABSTRACT

Glycosylation processes are under high natural selection pressure, presumably because these can modulate resistance to infection. Here, we asked whether inactivation of the UDP-galactose:ß-galactoside-α1-3-galactosyltransferase (α1,3GT) gene, which ablated the expression of the Galα1-3Galß1-4GlcNAc-R (α-gal) glycan and allowed for the production of anti-α-gal antibodies (Abs) in humans, confers protection against Plasmodium spp. infection, the causative agent of malaria and a major driving force in human evolution. We demonstrate that both Plasmodium spp. and the human gut pathobiont E. coli O86:B7 express α-gal and that anti-α-gal Abs are associated with protection against malaria transmission in humans as well as in α1,3GT-deficient mice, which produce protective anti-α-gal Abs when colonized by E. coli O86:B7. Anti-α-gal Abs target Plasmodium sporozoites for complement-mediated cytotoxicity in the skin, immediately after inoculation by Anopheles mosquitoes. Vaccination against α-gal confers sterile protection against malaria in mice, suggesting that a similar approach may reduce malaria transmission in humans.


Subject(s)
Escherichia coli/physiology , Immunoglobulin M/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/transmission , Plasmodium/physiology , Polysaccharides/immunology , Adult , Animals , Anopheles/parasitology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Autoantigens/immunology , Cell Line, Tumor , Child , Escherichia coli/classification , Escherichia coli/immunology , Female , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Gastrointestinal Tract/microbiology , Germ-Free Life , Humans , Immunoglobulin M/blood , Malaria, Falciparum/microbiology , Malaria, Falciparum/parasitology , Mice , Plasmodium/classification , Plasmodium/growth & development , Plasmodium/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/physiology , Sporozoites/immunology , Toll-Like Receptor 9/agonists
3.
Immunity ; 51(4): 750-765.e10, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31492649

ABSTRACT

Immunity that controls parasitemia and inflammation during Plasmodium falciparum (Pf) malaria can be acquired with repeated infections. A limited understanding of this complex immune response impedes the development of vaccines and adjunctive therapies. We conducted a prospective systems biology study of children who differed in their ability to control parasitemia and fever following Pf infection. By integrating whole-blood transcriptomics, flow-cytometric analysis, and plasma cytokine and antibody profiles, we demonstrate that a pre-infection signature of B cell enrichment, upregulation of T helper type 1 (Th1) and Th2 cell-associated pathways, including interferon responses, and p53 activation associated with control of malarial fever and coordinated with Pf-specific immunoglobulin G (IgG) and Fc receptor activation to control parasitemia. Our hypothesis-generating approach identified host molecules that may contribute to differential clinical outcomes during Pf infection. As a proof of concept, we have shown that enhanced p53 expression in monocytes attenuated Plasmodium-induced inflammation and predicted protection from fever.


Subject(s)
B-Lymphocytes/immunology , Blood Proteins/metabolism , Inflammation/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Tumor Suppressor Protein p53/metabolism , Adolescent , Adult , Animals , Antibodies, Protozoan/metabolism , Child , Child, Preschool , Disease Resistance , Female , Gene Expression Profiling , Humans , Infant , Interferons/metabolism , Male , Mice , Mice, Inbred C57BL , Prospective Studies , Receptors, Fc/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Young Adult
4.
N Engl J Med ; 390(17): 1549-1559, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38669354

ABSTRACT

BACKGROUND: Subcutaneous administration of the monoclonal antibody L9LS protected adults against controlled Plasmodium falciparum infection in a phase 1 trial. Whether a monoclonal antibody administered subcutaneously can protect children from P. falciparum infection in a region where this organism is endemic is unclear. METHODS: We conducted a phase 2 trial in Mali to assess the safety and efficacy of subcutaneous administration of L9LS in children 6 to 10 years of age over a 6-month malaria season. In part A of the trial, safety was assessed at three dose levels in adults, followed by assessment at two dose levels in children. In part B of the trial, children were randomly assigned, in a 1:1:1 ratio, to receive 150 mg of L9LS, 300 mg of L9LS, or placebo. The primary efficacy end point, assessed in a time-to-event analysis, was the first P. falciparum infection, as detected on blood smear performed at least every 2 weeks for 24 weeks. A secondary efficacy end point was the first episode of clinical malaria, as assessed in a time-to-event analysis. RESULTS: No safety concerns were identified in the dose-escalation part of the trial (part A). In part B, 225 children underwent randomization, with 75 children assigned to each group. No safety concerns were identified in part B. P. falciparum infection occurred in 36 participants (48%) in the 150-mg group, in 30 (40%) in the 300-mg group, and in 61 (81%) in the placebo group. The efficacy of L9LS against P. falciparum infection, as compared with placebo, was 66% (adjusted confidence interval [95% CI], 45 to 79) with the 150-mg dose and 70% (adjusted 95% CI, 50 to 82) with the 300-mg dose (P<0.001 for both comparisons). Efficacy against clinical malaria was 67% (adjusted 95% CI, 39 to 82) with the 150-mg dose and 77% (adjusted 95% CI, 55 to 89) with the 300-mg dose (P<0.001 for both comparisons). CONCLUSIONS: Subcutaneous administration of L9LS to children was protective against P. falciparum infection and clinical malaria over a period of 6 months. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT05304611.).


Subject(s)
Antibodies, Monoclonal, Humanized , Malaria, Falciparum , Adult , Child , Female , Humans , Male , Dose-Response Relationship, Drug , Double-Blind Method , Endemic Diseases/prevention & control , Injections, Subcutaneous , Kaplan-Meier Estimate , Malaria, Falciparum/drug therapy , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Mali/epidemiology , Plasmodium falciparum , Treatment Outcome , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Directly Observed Therapy , Artemether, Lumefantrine Drug Combination/administration & dosage , Artemether, Lumefantrine Drug Combination/therapeutic use , Young Adult , Middle Aged
5.
Immunity ; 45(2): 402-14, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27473412

ABSTRACT

Humoral immunity consists of pre-existing antibodies expressed by long-lived plasma cells and rapidly reactive memory B cells (MBC). Recent studies of MBC development and function after protein immunization have uncovered significant MBC heterogeneity. To clarify functional roles for distinct MBC subsets during malaria infection, we generated tetramers that identify Plasmodium-specific MBCs in both humans and mice. Long-lived murine Plasmodium-specific MBCs consisted of three populations: somatically hypermutated immunoglobulin M(+) (IgM(+)) and IgG(+) MBC subsets and an unmutated IgD(+) MBC population. Rechallenge experiments revealed that high affinity, somatically hypermutated Plasmodium-specific IgM(+) MBCs proliferated and gave rise to antibody-secreting cells that dominated the early secondary response to parasite rechallenge. IgM(+) MBCs also gave rise to T cell-dependent IgM(+) and IgG(+)B220(+)CD138(+) plasmablasts or T cell-independent B220(-)CD138(+) IgM(+) plasma cells. Thus, even in competition with IgG(+) MBCs, IgM(+) MBCs are rapid, plastic, early responders to a secondary Plasmodium rechallenge and should be targeted by vaccine strategies.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria/immunology , Plasmodium/immunology , Animals , Antibody Affinity , Antigens, Protozoan/immunology , Cells, Cultured , Epitopes , Female , Humans , Immunity, Humoral , Immunization, Secondary , Immunoglobulin M/metabolism , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Somatic Hypermutation, Immunoglobulin , T-Lymphocytes/immunology
6.
Malar J ; 22(1): 42, 2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36737743

ABSTRACT

BACKGROUND: In malaria endemic regions, transmission of Plasmodium falciparum parasites is often seasonal with very low transmission during the dry season and high transmission in the wet season. Parasites survive the dry season within some individuals who experience prolonged carriage of parasites and are thought to 'seed' infection in the next transmission season. METHODS: Dry season carriers and their role in the subsequent transmission season are characterized using a combination of mathematical simulations and data analysis of previously described data from a longitudinal study in Mali of individuals aged 3 months-12 years (n = 579). RESULTS: Simulating the life-history of individuals experiencing repeated exposure to infection predicts that dry season carriage is more likely in the oldest, most exposed and most immune individuals. This hypothesis is supported by the data from Mali, which shows that carriers are significantly older, experience a higher biting rate at the beginning of the transmission season and develop clinical malaria later than non-carriers. Further, since the most exposed individuals in a community are most likely to be dry season carriers, this is predicted to enable a more than twofold faster spread of parasites into the mosquito population at the start of the subsequent wet season. CONCLUSIONS: Carriage of malaria parasites over the months-long dry season in Mali is most likely in the older, more exposed and more immune children. These children may act as super-spreaders facilitating the fast spread of parasites at the beginning of the next transmission season.


Subject(s)
Malaria, Falciparum , Malaria , Parasites , Child , Animals , Humans , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Seasons , Longitudinal Studies , Plasmodium falciparum , Malaria/epidemiology
7.
PLoS Pathog ; 13(9): e1006576, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28953967

ABSTRACT

Many chronic infections, including malaria and HIV, are associated with a large expansion of CD21-CD27- 'atypical' memory B cells (MBCs) that exhibit reduced B cell receptor (BCR) signaling and effector functions. Little is known about the conditions or transcriptional regulators driving atypical MBC differentiation. Here we show that atypical MBCs in malaria-exposed individuals highly express the transcription factor T-bet, and that T-bet expression correlates inversely with BCR signaling and skews toward IgG3 class switching. Moreover, a longitudinal analysis of a subset of children suggested a correlation between the incidence of febrile malaria and the expansion of T-bethi B cells. The Th1-cytokine containing supernatants of malaria-stimulated PBMCs plus BCR cross linking induced T-bet expression in naïve B cells that was abrogated by neutralizing IFN-γ or blocking the IFN-γ receptor on B cells. Accordingly, recombinant IFN-γ plus BCR cross-linking drove T-bet expression in peripheral and tonsillar B cells. Consistent with this, Th1-polarized Tfh (Tfh-1) cells more efficiently induced T-bet expression in naïve B cells. These data provide new insight into the mechanisms underlying atypical MBC differentiation.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Gene Expression Regulation/immunology , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Malaria/immunology , Adolescent , Adult , Child , Child, Preschool , Female , Fetal Proteins/metabolism , Humans , Infant , Malaria/metabolism , Male , Receptors, Antigen, B-Cell/metabolism , T-Box Domain Proteins/metabolism , Young Adult
9.
PLoS Pathog ; 12(12): e1006054, 2016 12.
Article in English | MEDLINE | ID: mdl-27926944

ABSTRACT

Malaria remains one of the greatest burdens to global health, causing nearly 500,000 deaths in 2014. When manifesting in the lungs, severe malaria causes acute lung injury/acute respiratory distress syndrome (ALI/ARDS). We have previously shown that a proportion of DBA/2 mice infected with Plasmodium berghei ANKA (PbA) develop ALI/ARDS and that these mice recapitulate various aspects of the human syndrome, such as pulmonary edema, hemorrhaging, pleural effusion and hypoxemia. Herein, we investigated the role of neutrophils in the pathogenesis of malaria-associated ALI/ARDS. Mice developing ALI/ARDS showed greater neutrophil accumulation in the lungs compared with mice that did not develop pulmonary complications. In addition, mice with ALI/ARDS produced more neutrophil-attracting chemokines, myeloperoxidase and reactive oxygen species. We also observed that the parasites Plasmodium falciparum and PbA induced the formation of neutrophil extracellular traps (NETs) ex vivo, which were associated with inflammation and tissue injury. The depletion of neutrophils, treatment with AMD3100 (a CXCR4 antagonist), Pulmozyme (human recombinant DNase) or Sivelestat (inhibitor of neutrophil elastase) decreased the development of malaria-associated ALI/ARDS and significantly increased mouse survival. This study implicates neutrophils and NETs in the genesis of experimentally induced malaria-associated ALI/ARDS and proposes a new therapeutic approach to improve the prognosis of severe malaria.


Subject(s)
Acute Lung Injury/immunology , Neutrophils/immunology , Respiratory Distress Syndrome/immunology , Acute Lung Injury/microbiology , Animals , Disease Models, Animal , Extracellular Traps/immunology , Fluorescent Antibody Technique , Malaria/complications , Malaria/immunology , Male , Mice , Mice, Inbred DBA , Polymerase Chain Reaction , Respiratory Distress Syndrome/microbiology
10.
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
11.
Cell Immunol ; 321: 18-25, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28735813

ABSTRACT

Immunological memory is a remarkable phenomenon in which survival of an initial infection by a pathogen leads to life-long protection from disease upon subsequent exposure to that same pathogen. For many infectious diseases, long-lived protective humoral immunity is induced after only a single infection in a process that depends on the generation of memory B cells (MBCs) and long-lived plasma cells. However, over the past decade it has become increasingly evident that many chronic human infectious diseases to which immunity is not readily established, including HIV-AIDS, malaria and TB, are associated with fundamental alterations in the composition and functionality of MBC compartments. A common feature of these diseases appears to be a large expansion of what have been termed exhausted B cells, tissue-like memory B cells or atypical memory B cells (aMBCs) that, for simplicity's sake, we refer to here as aMBCs. It has been suggested that chronic immune activation and inflammation drive the expansion of aMBCs and that in some way aMBCs contribute to deficiencies in the acquisition of immunity in chronic infectious diseases. Although aMBCs are heterogeneous both within individuals and between diseases, they have several features in common including low expression of the cell surface markers that define classical MBCs in humans including CD21 and CD27 and high expression of genes not usually expressed by classical MBCs including T-bet, CD11c and a variety of inhibitory receptors, notably members of the FcRL family. Another distinguishing feature is their greatly diminished ability to be stimulated through their B cell receptors to proliferate, secrete cytokines or produce antibodies. In this review, we describe our current understanding of the phenotypic markers of aMBCs, their specificity in relation to the disease-causing pathogen, their functionality, the drivers of their expansion in chronic infections and their life span. We briefly summarize the features of aMBCs in healthy individuals and in autoimmune disease. We also comment on the possible relationship of human aMBCs and T-bet+, CD11c+ age/autoimmune-associated B cells, also a topic of this review volume.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Communicable Diseases/immunology , Immunologic Memory/immunology , B-Lymphocytes/metabolism , Chronic Disease , HIV Infections/immunology , Humans , Malaria/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Tuberculosis/immunology
12.
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
13.
J Immunol ; 190(7): 3039-46, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23526829

ABSTRACT

Plasmodium falciparum malaria remains a major public health threat for which there is no licensed vaccine. Abs play a key role in malaria immunity, but Ab-mediated protection is only acquired after years of repeated infections, leaving children in endemic areas vulnerable to severe malaria and death. Many P. falciparum Ags are extraordinarily diverse and clonally variant, which likely contribute to the inefficient acquisition of protective Abs. However, mounting evidence suggests that there is more to the story and that infection-induced dysregulation of B cell function also plays a role. We herein review progress toward understanding the B cell biology of P. falciparum infection, focusing on what has been learned from population-based studies in malaria-endemic areas. We suggest ways in which advances in immunology and genomics-based technology can further improve our understanding of the B cell response in malaria and perhaps illuminate new pathways to the development of effective vaccines.


Subject(s)
Antibodies, Protozoan/immunology , B-Lymphocytes/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Age Factors , Humans , Immunoglobulin G/immunology , Immunologic Memory , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/growth & development
14.
Nat Med ; 13(6): 703-10, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17496899

ABSTRACT

Cerebral malaria claims more than 1 million lives per year. We report that heme oxygenase-1 (HO-1, encoded by Hmox1) prevents the development of experimental cerebral malaria (ECM). BALB/c mice infected with Plasmodium berghei ANKA upregulated HO-1 expression and activity and did not develop ECM. Deletion of Hmox1 and inhibition of HO activity increased ECM incidence to 83% and 78%, respectively. HO-1 upregulation was lower in infected C57BL/6 compared to BALB/c mice, and all infected C57BL/6 mice developed ECM (100% incidence). Pharmacological induction of HO-1 and exposure to the end-product of HO-1 activity, carbon monoxide (CO), reduced ECM incidence in C57BL/6 mice to 10% and 0%, respectively. Whereas neither HO-1 nor CO affected parasitemia, both prevented blood-brain barrier (BBB) disruption, brain microvasculature congestion and neuroinflammation, including CD8(+) T-cell brain sequestration. These effects were mediated by the binding of CO to hemoglobin, preventing hemoglobin oxidation and the generation of free heme, a molecule that triggers ECM pathogenesis.


Subject(s)
Carbon Monoxide/physiology , Heme Oxygenase-1/physiology , Heme/metabolism , Malaria, Cerebral/enzymology , Animals , Disease Models, Animal , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Plasmodium berghei
15.
Nat Commun ; 15(1): 1413, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360754

ABSTRACT

Genomic surveillance of Plasmodium falciparum malaria can provide policy-relevant information about antimalarial drug resistance, diagnostic test failure, and the evolution of vaccine targets. Yet the large and low complexity genome of P. falciparum complicates the development of genomic methods, while resource constraints in malaria endemic regions can limit their deployment. Here, we demonstrate an approach for targeted nanopore sequencing of P. falciparum from dried blood spots (DBS) that enables cost-effective genomic surveillance of malaria in low-resource settings. We release software that facilitates flexible design of amplicon sequencing panels and use this software to design two target panels for P. falciparum. The panels generate 3-4 kbp reads for eight and sixteen targets respectively, covering key drug-resistance associated genes, diagnostic test antigens, polymorphic markers and the vaccine target csp. We validate our approach on mock and field samples, demonstrating robust sequencing coverage, accurate variant calls within coding sequences, the ability to explore P. falciparum within-sample diversity and to detect deletions underlying rapid diagnostic test failure.


Subject(s)
Malaria, Falciparum , Malaria , Nanopore Sequencing , Vaccines , Humans , Plasmodium falciparum/genetics , Cost-Benefit Analysis , Malaria, Falciparum/diagnosis , Malaria/epidemiology , Genomics
16.
EMBO Rep ; 12(12): 1233-42, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22081142

ABSTRACT

After the bite of a malaria-infected mosquito, the Plasmodium sporozoite infects liver cells and produces thousands of merozoites, which then infect red blood cells, causing malaria. In malaria-endemic areas, several hundred infected mosquitoes can bite an individual each year, increasing the risk of superinfection. However, in infants that are yet to acquire immunity, superinfections are infrequent. We have recently shown that blood-stage parasitaemia, above a minimum threshold, impairs the growth of a subsequent sporozoite infection of liver cells. Blood-stage parasites stimulate the production of the host iron-regulatory factor hepcidin, which redistributes iron away from hepatocytes, reducing the development of the iron-dependent liver stage. This could explain why Plasmodium superinfection is not often found in young nonimmune children. Here, we discuss the impact that such protection from superinfection might have in epidemiological settings or in programmes for controlling malaria, as well as how the induction of hepcidin and redistribution of iron might influence anaemia and the outcome of non-Plasmodium co-infections.


Subject(s)
Iron/metabolism , Malaria/complications , Malaria/parasitology , Plasmodium/physiology , Superinfection/complications , Superinfection/parasitology , Host-Parasite Interactions/immunology , Humans , Immunity , Malaria/prevention & control , Plasmodium/growth & development
17.
Malar J ; 11: 162, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22577737

ABSTRACT

BACKGROUND: Several studies indicate that people of the Fulani ethnic group are less susceptible to malaria compared to those of other ethnic groups living sympatrically in Africa, including the Dogon ethnic group. Although the mechanisms of this protection remain unclear, the Fulani are known to have higher levels of Plasmodium falciparum-specific antibodies of all Ig classes as compared to the Dogon. However, the proportions of B cell subsets in the Fulani and Dogon that may account for differences in the levels of Ig have not been characterized. METHODS: In this cross-sectional study, venous blood was collected from asymptomatic Fulani (n = 25) and Dogon (n = 25) adults in Mali during the malaria season, and from P. falciparum-naïve adults in the U.S. (n = 8). At the time of the blood collection, P. falciparum infection was detected by blood-smear in 16% of the Fulani and 36% of the Dogon volunteers. Thawed lymphocytes were analysed by flow cytometry to quantify B cell subsets, including immature and naïve B cells; plasma cells; and classical, activated, and atypical memory B cells (MBCs). RESULTS: The overall distribution of B cell subsets was similar between Fulani and Dogon adults, although the percentage of activated MBCs was higher in the Fulani group (Fulani: 11.07% [95% CI: 9.317 - 12.82]; Dogon: 8.31% [95% CI: 6.378 - 10.23]; P = 0.016). The percentage of atypical MBCs was similar between Fulani and Dogon adults (Fulani: 28.3% [95% CI: 22.73 - 34.88]; Dogon: 29.3% [95% CI: 25.06 - 33.55], but higher than U.S. adults (U.S.: 3.0% [95% CI: -0.21 - 6.164]; P < 0.001). Plasmodium falciparum infection was associated with a higher percentage of plasma cells among Fulani (Fulani infected: 3.3% [95% CI: 1.788 - 4.744]; Fulani uninfected: 1.71% [95% CI: 1.33 - 2.08]; P = 0.011), but not Dogon adults. CONCLUSION: These data show that the malaria-resistant Fulani have a higher percentage of activated MBCs compared to the Dogon, and that P. falciparum infection is associated with a higher percentage of plasma cells in the Fulani compared to the Dogon, findings that may account for the higher levels of P. falciparum antibodies in the Fulani.


Subject(s)
B-Lymphocytes/immunology , Disease Susceptibility , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adult , Cross-Sectional Studies , Ethnicity , Female , Flow Cytometry , Humans , Leukocyte Count , Lymphocyte Subsets/immunology , Male , Mali , Middle Aged , United States , Young Adult
18.
Mol Ecol Resour ; 22(6): 2285-2303, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35437908

ABSTRACT

Multiplexed PCR amplicon sequencing (AmpSeq) is an increasingly popular application for cost-effective monitoring of threatened species and managed wildlife populations, and shows strong potential for the genomic epidemiology of infectious disease. AmpSeq data from infectious microbes can inform disease control in multiple ways, such as by measuring drug resistance marker prevalence, distinguishing imported from local cases, and determining the effectiveness of therapeutics. We describe the design and comparative evaluation of two new AmpSeq assays for Plasmodium falciparum malaria parasites: a four-locus panel ("4CAST") composed of highly diverse antigens, and a 129-locus panel ("AMPLseq") composed of drug resistance markers, highly diverse loci for inferring relatedness, and a locus to detect Plasmodium vivax co-infection. We explore the performance of each panel in various public health use cases with in silico simulations as well as empirical experiments. The 4CAST panel appears highly suitable for evaluating the number of distinct parasite strains within samples (complexity of infection), showing strong performance across a wide range of parasitaemia levels without a DNA pre-amplification step. For relatedness inference, the larger AMPLseq panel performs similarly to two existing panels of comparable size, despite differences in the data and approach used for designing each panel. Finally, we describe an R package (paneljudge) that facilitates the design and comparative evaluation of genetic panels for relatedness estimation, and we provide general guidance on the design and implementation of AmpSeq panels for the genomic epidemiology of infectious disease.


Subject(s)
Communicable Diseases , Malaria, Vivax , Malaria , Genomics , Humans , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Plasmodium falciparum/genetics , Plasmodium vivax/genetics
19.
Nat Commun ; 12(1): 4711, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34330920

ABSTRACT

Following Plasmodium falciparum infection, individuals can remain asymptomatic, present with mild fever in uncomplicated malaria cases, or show one or more severe malaria symptoms. Several studies have investigated associations between parasite transcription and clinical severity, but no broad conclusions have yet been drawn. Here, we apply a series of bioinformatic approaches based on P. falciparum's tightly regulated transcriptional pattern during its ~48-hour intraerythrocytic developmental cycle (IDC) to publicly available transcriptomes of parasites obtained from malaria cases of differing clinical severity across multiple studies. Our analysis shows that within each IDC, the circulation time of infected erythrocytes without sequestering to endothelial cells decreases with increasing parasitaemia or disease severity. Accordingly, we find that the size of circulating infected erythrocytes is inversely related to parasite density and disease severity. We propose that enhanced adhesiveness of infected erythrocytes leads to a rapid increase in parasite burden, promoting higher parasitaemia and increased disease severity.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Bacterial , Malaria, Falciparum/blood , Parasitemia/blood , Plasmodium falciparum/genetics , Blood Circulation Time , Erythrocytes/parasitology , Gene Ontology , Genes, Bacterial/genetics , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/physiopathology , Parasitemia/parasitology , Parasitemia/physiopathology , Plasmodium falciparum/physiology
20.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33661302

ABSTRACT

Multimeric immunoglobulin-like molecules arose early in vertebrate evolution, yet the unique contributions of multimeric IgM antibodies to infection control are not well understood. This is partially due to the difficulty of distinguishing low-affinity IgM, secreted rapidly by plasmablasts, from high-affinity antibodies derived from later-arising memory cells. We developed a pipeline to express B cell receptors (BCRs) from Plasmodium falciparum-specific IgM+ and IgG+ human memory B cells (MBCs) as both IgM and IgG molecules. BCRs from both subsets were somatically hypermutated and exhibited comparable monomeric affinity. Crystallization of one IgM+ MBC-derived antibody complexed with antigen defined a linear epitope within a conserved Plasmodium protein. In its physiological multimeric state, this antibody displayed exponentially higher antigen binding than a clonally identical IgG monomer, and more effectively inhibited P. falciparum invasion. Forced multimerization of this IgG significantly improved both antigen binding and parasite restriction, underscoring how avidity can alter antibody function. This work demonstrates the potential of high-avidity IgM in both therapeutics and vaccines.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , B-Lymphocytes/immunology , Immunoglobulin M/chemistry , Immunoglobulin M/immunology , Immunologic Memory , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protein Multimerization/immunology , Adolescent , Antibody Affinity , Cells, Cultured , Child , Cohort Studies , Epitopes, B-Lymphocyte/immunology , Female , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Male , Mali , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology
SELECTION OF CITATIONS
SEARCH DETAIL