Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 595(7865): 96-100, 2021 07.
Article in English | MEDLINE | ID: mdl-34040257

ABSTRACT

Trypanosomes are protozoan parasites that cause infectious diseases, including African trypanosomiasis (sleeping sickness) in humans and nagana in economically important livestock1,2. An effective vaccine against trypanosomes would be an important control tool, but the parasite has evolved sophisticated immunoprotective mechanisms-including antigenic variation3-that present an apparently insurmountable barrier to vaccination. Here we show, using a systematic genome-led vaccinology approach and a mouse model of Trypanosoma vivax infection4, that protective invariant subunit vaccine antigens can be identified. Vaccination with a single recombinant protein comprising the extracellular region of a conserved cell-surface protein that is localized to the flagellum membrane (which we term 'invariant flagellum antigen from T. vivax') induced long-lasting protection. Immunity was passively transferred with immune serum, and recombinant monoclonal antibodies to this protein could induce sterile protection and revealed several mechanisms of antibody-mediated immunity, including a major role for complement. Our discovery identifies a vaccine candidate for an important parasitic disease that has constrained socioeconomic development in countries in sub-Saharan Africa5, and provides evidence that highly protective vaccines against trypanosome infections can be achieved.


Subject(s)
Antigens, Protozoan/immunology , Protozoan Vaccines/immunology , Trypanosoma vivax/immunology , Trypanosomiasis, African/immunology , Trypanosomiasis, African/prevention & control , Animals , Antigens, Protozoan/chemistry , Complement System Proteins/immunology , Conserved Sequence/immunology , Disease Models, Animal , Female , Flagella/chemistry , Flagella/immunology , Mice , Mice, Inbred BALB C , Protozoan Vaccines/chemistry , Time Factors , Trypanosoma vivax/chemistry , Trypanosoma vivax/cytology , Trypanosomiasis, African/parasitology , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology
2.
Proc Natl Acad Sci U S A ; 121(5): e2316304121, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38261617

ABSTRACT

The discovery that Africans were resistant to infection by Plasmodium vivax (P. vivax) led to the conclusion that P. vivax invasion relied on the P. vivax Duffy Binding Protein (PvDBP) interacting with the Duffy Antigen Receptor for Chemokines (DARC) expressed on erythrocytes. However, the recent reporting of P. vivax infections in DARC-negative Africans suggests that the parasite might use an alternate invasion pathway to infect DARC-negative reticulocytes. To identify the parasite ligands and erythrocyte receptors that enable P. vivax invasion of both DARC-positive and -negative erythrocytes, we expressed region II containing the Duffy Binding-Like (DBL) domain of P. vivax erythrocyte binding protein (PvEBP-RII) and verified that the DBL domain binds to both DARC-positive and -negative erythrocytes. Furthermore, an AVidity-based EXtracelluar Interaction Screening (AVEXIS) was used to identify the receptor for PvEBP among over 750 human cell surface receptor proteins, and this approach identified only Complement Receptor 1 (CR1, CD35, or C3b/C4b receptor) as a PvEBP receptor. CR1 is a well-known receptor for P. falciparum Reticulocyte binding protein Homology 4 (PfRh4) and is present on the surfaces of both reticulocytes and normocytes, but its expression decreases as erythrocytes age. Indeed, PvEBP-RII bound to a subpopulation of both reticulocytes and normocytes, and this binding was blocked by the addition of soluble CR1 recombinant protein, indicating that CR1 is the receptor of PvEBP. In addition, we found that the Long Homology Repeat A (LHR-A) subdomain of CR1 is the only subdomain responsible for mediating the interaction with PvEBP-RII.


Subject(s)
Malaria, Falciparum , Plasmodium vivax , Humans , Receptors, Cell Surface , Erythrocytes , Reticulocytes , CD2 Antigens , Cell Adhesion Molecules
3.
J Infect Dis ; 225(8): 1435-1446, 2022 04 19.
Article in English | MEDLINE | ID: mdl-32524140

ABSTRACT

BACKGROUND: Schistosomiasis is a major global health problem caused by blood-dwelling parasitic worms, which is currently tackled primarily by mass administration of the drug praziquantel. Appropriate drug treatment strategies are informed by diagnostics that establish the prevalence and intensity of infection, which, in regions of low transmission, should be highly sensitive. METHODS: To identify sensitive new serological markers of Schistosoma mansoni infections, we have compiled a recombinant protein library of parasite cell-surface and secreted proteins expressed in mammalian cells. RESULTS: Together with a time series of sera samples from volunteers experimentally infected with a defined number of male parasites, we probed this protein library to identify several markers that can detect primary infections with as low as 10 parasites and as early as 5 weeks postinfection. CONCLUSIONS: These new markers could be further explored as valuable tools to detect ongoing and previous S mansoni infections, including in endemic regions where transmission is low.


Subject(s)
Schistosomiasis mansoni , Schistosomiasis , Animals , Biomarkers , Humans , Male , Mammals , Mice , Praziquantel/therapeutic use , Recombinant Proteins , Schistosoma mansoni , Schistosomiasis/drug therapy , Schistosomiasis mansoni/diagnosis , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/parasitology
4.
Proc Natl Acad Sci U S A ; 115(17): 4477-4482, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29632205

ABSTRACT

Malaria-causing Plasmodium sporozoites are deposited in the dermis by the bite of an infected mosquito and move by gliding motility to the liver where they invade and develop within host hepatocytes. Although extracellular interactions between Plasmodium sporozoite ligands and host receptors provide important guidance cues for productive infection and are good vaccine targets, these interactions remain largely uncharacterized. Thrombospondin-related anonymous protein (TRAP) is a parasite cell surface ligand that is essential for both gliding motility and invasion because it couples the extracellular binding of host receptors to the parasite cytoplasmic actinomyosin motor; however, the molecular nature of the host TRAP receptors is poorly defined. Here, we use a systematic extracellular protein interaction screening approach to identify the integrin αvß3 as a directly interacting host receptor for Plasmodium falciparum TRAP. Biochemical characterization of the interaction suggests a two-site binding model, requiring contributions from both the von Willebrand factor A domain and the RGD motif of TRAP for integrin binding. We show that TRAP binding to cells is promoted in the presence of integrin-activating proadhesive Mn2+ ions, and that cells genetically targeted so that they lack cell surface expression of the integrin αv-subunit are no longer able to bind TRAP. P. falciparum sporozoites moved with greater speed in the dermis of Itgb3-deficient mice, suggesting that the interaction has a role in sporozoite migration. The identification of the integrin αvß3 as the host receptor for TRAP provides an important demonstration of a sporozoite surface ligand that directly interacts with host receptors.


Subject(s)
Integrin alphaVbeta3/metabolism , Models, Biological , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Sporozoites/metabolism , Animals , HEK293 Cells , Humans , Integrin alphaVbeta3/genetics , Mice , Mice, Knockout , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protozoan Proteins/genetics , Sporozoites/genetics
5.
Proc Natl Acad Sci U S A ; 114(45): 12045-12050, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078270

ABSTRACT

A highly effective vaccine would be a valuable weapon in the drive toward malaria elimination. No such vaccine currently exists, and only a handful of the hundreds of potential candidates in the parasite genome have been evaluated. In this study, we systematically evaluated 29 antigens likely to be involved in erythrocyte invasion, an essential developmental stage during which the malaria parasite is vulnerable to antibody-mediated inhibition. Testing antigens alone and in combination identified several strain-transcending targets that had synergistic combinatorial effects in vitro, while studies in an endemic population revealed that combinations of the same antigens were associated with protection from febrile malaria. Video microscopy established that the most effective combinations targeted multiple discrete stages of invasion, suggesting a mechanistic explanation for synergy. Overall, this study both identifies specific antigen combinations for high-priority clinical testing and establishes a generalizable approach that is more likely to produce effective vaccines.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Antibodies, Protozoan/immunology , Cell Line , Erythrocytes/immunology , Erythrocytes/parasitology , HEK293 Cells , Humans , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Prospective Studies , Protozoan Proteins/immunology
6.
J Biol Chem ; 291(27): 14285-14299, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27226583

ABSTRACT

Diversity at pathogen genetic loci can be driven by host adaptive immune selection pressure and may reveal proteins important for parasite biology. Population-based genome sequencing of Plasmodium falciparum, the parasite responsible for the most severe form of malaria, has highlighted two related polymorphic genes called dblmsp and dblmsp2, which encode Duffy binding-like (DBL) domain-containing proteins located on the merozoite surface but whose function remains unknown. Using recombinant proteins and transgenic parasites, we show that DBLMSP and DBLMSP2 directly and avidly bind human IgM via their DBL domains. We used whole genome sequence data from over 400 African and Asian P. falciparum isolates to show that dblmsp and dblmsp2 exhibit extreme protein polymorphism in their DBL domain, with multiple variants of two major allelic classes present in every population tested. Despite this variability, the IgM binding function was retained across diverse sequence representatives. Although this interaction did not seem to have an effect on the ability of the parasite to invade red blood cells, binding of DBLMSP and DBLMSP2 to IgM inhibited the overall immunoreactivity of these proteins to IgG from patients who had been exposed to the parasite. This suggests that IgM binding might mask these proteins from the host humoral immune system.


Subject(s)
Antigens, Protozoan/metabolism , Immunoglobulin M/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Humans , Protein Binding
7.
Nature ; 480(7378): 534-7, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22080952

ABSTRACT

Erythrocyte invasion by Plasmodium falciparum is central to the pathogenesis of malaria. Invasion requires a series of extracellular recognition events between erythrocyte receptors and ligands on the merozoite, the invasive form of the parasite. None of the few known receptor-ligand interactions involved are required in all parasite strains, indicating that the parasite is able to access multiple redundant invasion pathways. Here, we show that we have identified a receptor-ligand pair that is essential for erythrocyte invasion in all tested P. falciparum strains. By systematically screening a library of erythrocyte proteins, we have found that the Ok blood group antigen, basigin, is a receptor for PfRh5, a parasite ligand that is essential for blood stage growth. Erythrocyte invasion was potently inhibited by soluble basigin or by basigin knockdown, and invasion could be completely blocked using low concentrations of anti-basigin antibodies; importantly, these effects were observed across all laboratory-adapted and field strains tested. Furthermore, Ok(a-) erythrocytes, which express a basigin variant that has a weaker binding affinity for PfRh5, had reduced invasion efficiencies. Our discovery of a cross-strain dependency on a single extracellular receptor-ligand pair for erythrocyte invasion by P. falciparum provides a focus for new anti-malarial therapies.


Subject(s)
Basigin/metabolism , Erythrocytes/parasitology , Host-Parasite Interactions , Plasmodium falciparum/physiology , Basigin/chemistry , Basigin/genetics , Erythrocytes/metabolism , Gene Knockdown Techniques , Humans , Models, Molecular , Protein Structure, Tertiary
8.
J Immunol ; 192(1): 245-58, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24293631

ABSTRACT

There is intense interest in induction and characterization of strain-transcending neutralizing Ab against antigenically variable human pathogens. We have recently identified the human malaria parasite Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) as a target of broadly neutralizing Abs, but there is little information regarding the functional mechanism(s) of Ab-mediated neutralization. In this study, we report that vaccine-induced polyclonal anti-PfRH5 Abs inhibit the tight attachment of merozoites to erythrocytes and are capable of blocking the interaction of PfRH5 with its receptor basigin. Furthermore, by developing anti-PfRH5 mAbs, we provide evidence of the following: 1) the ability to block the PfRH5-basigin interaction in vitro is predictive of functional activity, but absence of blockade does not predict absence of functional activity; 2) neutralizing mAbs bind spatially related epitopes on the folded protein, involving at least two defined regions of the PfRH5 primary sequence; 3) a brief exposure window of PfRH5 is likely to necessitate rapid binding of Ab to neutralize parasites; and 4) intact bivalent IgG contributes to but is not necessary for parasite neutralization. These data provide important insight into the mechanisms of broadly neutralizing anti-malaria Abs and further encourage anti-PfRH5-based malaria prevention efforts.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Carrier Proteins/immunology , Merozoites/immunology , Plasmodium falciparum/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antibodies, Protozoan/metabolism , Carrier Proteins/metabolism , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Kinetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Mice , Neutralization Tests , Plasmodium falciparum/growth & development , Protein Binding/immunology , Rabbits
9.
Nat Genet ; 39(3): 397-402, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17259985

ABSTRACT

During organogenesis, the foregut endoderm gives rise to the many different cell types that comprise the hepatopancreatic system, including hepatic, pancreatic and gallbladder cells, as well as the epithelial cells of the hepatopancreatic ductal system that connects these organs together and with the intestine. However, the mechanisms responsible for demarcating ducts versus organs are poorly understood. Here, we show that Fgf10 signaling from the adjacent mesenchyme is responsible for refining the boundaries between the hepatopancreatic duct and organs. In zebrafish fgf10 mutants, the hepatopancreatic ductal epithelium is severely dysmorphic, and cells of the hepatopancreatic ductal system and adjacent intestine misdifferentiate toward hepatic and pancreatic fates. Furthermore, Fgf10 also functions to prevent the differentiation of the proximal pancreas and liver into hepatic and pancreatic cells, respectively. These data shed light onto how the multipotent cells of the foregut endoderm, and subsequently those of the hepatopancreatic duct, are directed toward different organ fates.


Subject(s)
Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factors/metabolism , Hepatopancreas/embryology , Mesoderm/cytology , Organogenesis , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Body Patterning , Cell Differentiation , Embryo, Nonmammalian , Fibroblast Growth Factor 10/genetics , Fluorescent Antibody Technique , Hepatopancreas/anatomy & histology , Hepatopancreas/metabolism , Mesoderm/metabolism , Signal Transduction , Zebrafish/metabolism , Zebrafish Proteins/genetics
10.
Mol Cell Proteomics ; 12(12): 3976-86, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24043421

ABSTRACT

Malaria, an infectious disease caused by parasites of the Plasmodium genus, is one of the world's major public health concerns causing up to a million deaths annually, mostly because of P. falciparum infections. All of the clinical symptoms are associated with the blood stage of the disease, an obligate part of the parasite life cycle, when a form of the parasite called the merozoite recognizes and invades host erythrocytes. During erythrocyte invasion, merozoites are directly exposed to the host humoral immune system making the blood stage of the parasite a conceptually attractive therapeutic target. Progress in the functional and molecular characterization of P. falciparum merozoite proteins, however, has been hampered by the technical challenges associated with expressing these proteins in a biochemically active recombinant form. This challenge is particularly acute for extracellular proteins, which are the likely targets of host antibody responses, because they contain structurally critical post-translational modifications that are not added by some recombinant expression systems. Here, we report the development of a method that uses a mammalian expression system to compile a protein resource containing the entire ectodomains of 42 P. falciparum merozoite secreted and cell surface proteins, many of which have not previously been characterized. Importantly, we are able to recapitulate known biochemical activities by showing that recombinant MSP1-MSP7 and P12-P41 directly interact, and that both recombinant EBA175 and EBA140 can bind human erythrocytes in a sialic acid-dependent manner. Finally, we use sera from malaria-exposed immune adults to profile the relative immunoreactivity of the proteins and show that the majority of the antigens contain conformational (heat-labile) epitopes. We envisage that this resource of recombinant proteins will make a valuable contribution toward a molecular understanding of the blood stage of P. falciparum infections and facilitate the comparative screening of antigens as blood-stage vaccine candidates.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Surface/genetics , Erythrocytes/parasitology , Merozoites/metabolism , Plasmodium falciparum/genetics , Proteome/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Antigens, Surface/immunology , Antigens, Surface/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cloning, Molecular , Gene Expression , HEK293 Cells , Humans , Immune Sera/chemistry , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/immunology , Merozoite Surface Protein 1/metabolism , Merozoites/chemistry , Merozoites/immunology , Molecular Sequence Annotation , Peptide Library , Plasmodium falciparum/immunology , Plasmodium falciparum/metabolism , Protein Binding , Proteome/immunology , Proteome/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sialic Acids/metabolism
11.
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
12.
J Biol Chem ; 288(45): 32106-32117, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24043627

ABSTRACT

PfEBA175 has an important role in the invasion of human erythrocytes by Plasmodium falciparum and is therefore considered a high priority blood-stage malaria vaccine candidate. PfEBA175 mediates adhesion to erythrocytes through binding of the Duffy-binding-like (DBL) domains in its extracellular domain to Neu5Acα2-3Gal displayed on the O-linked glycans of glycophorin-A (GYPA). Because of the difficulties in expressing active full-length (FL) P. falciparum proteins in a recombinant form, previous analyses of the PfEBA175-GYPA interaction have largely focused on the DBL domains alone, and therefore they have not been performed in the context of the native protein sequence. Here, we express the entire ectodomain of PfEBA175 (PfEBA175 FL) in soluble form, allowing us to compare the biochemical and immunological properties with a fragment containing only the tandem DBL domains ("region II," PfEBA175 RII). Recombinant PfEBA175 FL bound human erythrocytes in a trypsin and neuraminidase-sensitive manner and recognized Neu5Acα2-3Gal-containing glycans, confirming its biochemical activity. A quantitative binding analysis showed that PfEBA175 FL interacted with native GYPA with a KD ∼0.26 µM and is capable of self-association. By comparison, the RII fragment alone bound GYPA with a lower affinity demonstrating that regions outside of the DBL domains are important for interactions with GYPA; antibodies directed to these other regions also contributed to the inhibition of parasite invasion. These data demonstrate the importance of PfEBA175 regions other than the DBL domains in the interaction with GYPA and merit their inclusion in an EBA175-based vaccine.


Subject(s)
Antigens, Protozoan/immunology , Erythrocytes/immunology , Glycophorins/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Glycophorins/genetics , Glycophorins/metabolism , Humans , Malaria Vaccines/genetics , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Malaria, Falciparum/prevention & control , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Proteins
13.
PLoS Pathog ; 8(11): e1003031, 2012.
Article in English | MEDLINE | ID: mdl-23166499

ABSTRACT

The motility and invasion of Plasmodium parasites is believed to require a cytoplasmic actin-myosin motor associated with a cell surface ligand belonging to the TRAP (thrombospondin-related anonymous protein) family. Current models of invasion usually invoke the existence of specific receptors for the TRAP-family ligands on the surface of the host cell; however, the identities of these receptors remain largely unknown. Here, we identify the GPI-linked protein Semaphorin-7A (CD108) as an erythrocyte receptor for the P. falciparum merozoite-specific TRAP homolog (MTRAP) by using a systematic screening approach designed to detect extracellular protein interactions. The specificity of the interaction was demonstrated by showing that binding was saturable and by quantifying the equilibrium and kinetic biophysical binding parameters using surface plasmon resonance. We found that two MTRAP monomers interact via their tandem TSR domains with the Sema domains of a Semaphorin-7A homodimer. Known naturally-occurring polymorphisms in Semaphorin-7A did not quantitatively affect MTRAP binding nor did the presence of glycans on the receptor. Attempts to block the interaction during in vitro erythrocyte invasion assays using recombinant proteins and antibodies showed no significant inhibitory effect, suggesting the inaccessibility of the complex to proteinaceous blocking agents. These findings now provide important experimental evidence to support the model that parasite TRAP-family ligands interact with specific host receptors during cellular invasion.


Subject(s)
Antigens, CD/metabolism , Erythrocytes/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Semaphorins/metabolism , Animals , Antigens, CD/genetics , Erythrocytes/parasitology , Erythrocytes/pathology , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Malaria, Falciparum/genetics , Malaria, Falciparum/pathology , Male , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Polymorphism, Genetic , Protein Multimerization/genetics , Protein Structure, Tertiary , Protozoan Proteins/genetics , Semaphorins/genetics
14.
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
15.
Cell Microbiol ; 15(8): 1304-12, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23617720

ABSTRACT

The invasion of host erythrocytes by the parasite Plasmodium falciparum initiates the blood stage of infection responsible for the symptoms of malaria. Invasion involves extracellular protein interactions between host erythrocyte receptors and ligands on the merozoite, the invasive form of the parasite. Despite significant research effort, many merozoite surface ligands have no known erythrocyte binding partner, most likely due to the intractable biochemical nature of membrane-tethered receptor proteins and their interactions. The few receptor-ligand pairs that have been described have largely relied on sourcing erythrocytes from patients with rare blood groups, a serendipitous approach that is unsatisfactory for systematically identifying novel receptors. We have recently developed a scalable assay called AVEXIS (for AVidity-based EXtracellular Interaction Screen), designed to circumvent the technical difficulties associated with the identification of extracellular protein interactions, and applied it to identify erythrocyte receptors for orphan P. falciparum merozoite ligands. Using this approach, we have recently identified Basigin (CD147) and Semaphorin-7A (CD108) as receptors for RH5 and MTRAP respectively. In this essay, we review techniques used to identify Plasmodium receptors and discuss how they could be applied in the future to identify novel receptors both for Plasmodium parasites but also other pathogens.


Subject(s)
Erythrocytes/parasitology , Plasmodium falciparum/metabolism , Protein Interaction Maps , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Basigin/metabolism , Carrier Proteins/metabolism , Humans , Merozoites/metabolism , Plasmodium falciparum/isolation & purification , Semaphorins/metabolism
16.
Phys Rev Lett ; 110(22): 220501, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23767703

ABSTRACT

We devise powerful algorithms based on differential evolution for adaptive many-particle quantum metrology. Our new approach delivers adaptive quantum metrology policies for feedback control that are orders-of-magnitude more efficient and surpass the few-dozen-particle limitation arising in methods based on particle-swarm optimization. We apply our method to the binary-decision-tree model for quantum-enhanced phase estimation as well as to a new problem: a decision tree for adaptive estimation of the unknown bias of a quantum coin in a quantum walk and show how this latter case can be realized experimentally.

17.
Curr Biol ; 18(20): 1565-71, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18951027

ABSTRACT

Emerging evidence indicates that paracrine signals from endothelial cells play a role in tissue differentiation and organ formation [1-3]. Here, we identify a novel role for endothelial cells in modulating hepatocyte polarization during liver organogenesis. We find that in zebrafish, the apical domain of the hepatocytes predicts the location of the intrahepatic biliary network. The refinement of hepatocyte polarization coincides with the invasion of endothelial cells into the liver, and these endothelial cells migrate along the maturing basal surface of the hepatocytes. Using genetic, pharmacological, and transplantation experiments, we provide evidence that endothelial cells influence the polarization of the adjacent hepatocytes. This influence of endothelial cells on hepatocytes is mediated at least in part by the cell-surface protein Heart of glass and contributes to the establishment of coordinately aligned hepatocyte apical membranes and evenly spaced intrahepatic conduits.


Subject(s)
Cell Polarity , Endothelial Cells/metabolism , Hepatocytes/metabolism , Liver/embryology , Zebrafish/embryology , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , Cell Membrane/metabolism , Cell Movement , Gene Expression Regulation, Developmental , Liver/cytology , Membrane Glycoproteins/metabolism , Organogenesis , Zebrafish/genetics , Zebrafish Proteins/metabolism
18.
BMC Biol ; 8: 76, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20525357

ABSTRACT

BACKGROUND: Monoclonal antibodies with high affinity and selectivity that work on wholemount fixed tissues are valuable reagents to the cell and developmental biologist, and yet isolating them remains a long and unpredictable process. Here we report a rapid and scalable method to select and express recombinant mouse monoclonal antibodies that are essentially equivalent to those secreted by parental IgG-isotype hybridomas. RESULTS: Increased throughput was achieved by immunizing mice with pools of antigens and cloning - from small numbers of hybridoma cells - the functionally rearranged light and heavy chains into a single expression plasmid. By immunizing with the ectodomains of zebrafish cell surface receptor proteins expressed in mammalian cells and screening for formalin-resistant epitopes, we selected antibodies that gave expected staining patterns on wholemount fixed zebrafish embryos. CONCLUSIONS: This method can be used to quickly select several high quality monoclonal antibodies from a single immunized mouse and facilitates their distribution using plasmids.


Subject(s)
Antibodies, Monoclonal/genetics , Cloning, Molecular/methods , Recombinant Proteins/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Cell Line , Cell Line, Tumor , Gene Expression , Humans , Hybridomas/immunology , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred BALB C , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Zebrafish/embryology , Zebrafish/immunology
19.
Wellcome Open Res ; 4: 159, 2019.
Article in English | MEDLINE | ID: mdl-31728414

ABSTRACT

Background: Schistosomiasis is a major parasitic disease affecting people living in tropical and sup-tropical areas. Transmission of the parasite has been reported in 78 countries, causing significant morbidity and around 200,000 deaths per year in endemic regions. The disease is currently managed by the mass-administration of praziquantel to populations at risk of infection; however, the reliance on a single drug raises the prospect of parasite resistance to the only treatment widely available. The development of an effective vaccine would be a more powerful method of control, but none currently exists and the identification of new immunogens that can elicit protective immune responses therefore remains a priority. Because of the complex nature of the parasite life cycle, identification of new vaccine candidates has mostly relied on the use of animal models and on a limited set of recombinant proteins. Methods: In this study, we have established an infrastructure for testing a large number of vaccine candidates in mice and used it to screen 96 cell-surface and secreted recombinant proteins from Schistosoma mansoni. This approach, using standardised immunisation and percutaneous infection protocols, allowed us to compare an extensive set of antigens in a systematic manner. Results: Although some vaccine candidates were associated with a statistically significant reduction in the number of eggs in the initial screens, these observations could not be repeated in subsequent challenges and none of the proteins studied were associated with a strongly protective effect against infection. Conclusions: Although no antigens individually induced reproducible and strongly protective effects using our vaccination regime, we have established the experimental infrastructures to facilitate large-scale systematic subunit vaccine testing for schistosomiasis in a murine infection model.

20.
Cell Host Microbe ; 17(1): 130-9, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25590760

ABSTRACT

Antigenic diversity has posed a critical barrier to vaccine development against the pathogenic blood-stage infection of the human malaria parasite Plasmodium falciparum. To date, only strain-specific protection has been reported by trials of such vaccines in nonhuman primates. We recently showed that P. falciparum reticulocyte binding protein homolog 5 (PfRH5), a merozoite adhesin required for erythrocyte invasion, is highly susceptible to vaccine-inducible strain-transcending parasite-neutralizing antibody. In vivo efficacy of PfRH5-based vaccines has not previously been evaluated. Here, we demonstrate that PfRH5-based vaccines can protect Aotus monkeys against a virulent vaccine-heterologous P. falciparum challenge and show that such protection can be achieved by a human-compatible vaccine formulation. Protection was associated with anti-PfRH5 antibody concentration and in vitro parasite-neutralizing activity, supporting the use of this in vitro assay to predict the in vivo efficacy of future vaccine candidates. These data suggest that PfRH5-based vaccines have potential to achieve strain-transcending efficacy in humans.


Subject(s)
Carrier Proteins/immunology , Immunity, Heterologous , Malaria Vaccines/immunology , Malaria/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Aotus trivirgatus , Disease Models, Animal , Female , Malaria/immunology , Malaria Vaccines/administration & dosage , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL