Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
1.
Nat Commun ; 14(1): 6391, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828011

ABSTRACT

Placental malaria is caused by Plasmodium falciparum-infected erythrocytes (IEs) adhering to chondroitin sulfate proteoglycans in placenta via VAR2CSA-type PfEMP1. Human pentameric immunoglobulin M (IgM) binds to several types of PfEMP1, including VAR2CSA via its Fc domain. Here, a 3.6 Å cryo-electron microscopy map of the IgM-VAR2CSA complex reveals that two molecules of VAR2CSA bind to the Cµ4 of IgM through their DBL3X and DBL5ε domains. The clockwise and anti-clockwise rotation of the two VAR2CSA molecules on opposite faces of IgM juxtaposes C-termini of both VAR2CSA near the J chain, where IgM creates a wall between both VAR2CSA molecules and hinders its interaction with its receptor. To support this, we show when VAR2CSA is bound to IgM, its staining on IEs as well as binding of IEs to chondroitin sulfate A in vitro is severely compromised.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Female , Pregnancy , Humans , Plasmodium falciparum/metabolism , Chondroitin Sulfates/metabolism , Cryoelectron Microscopy , Placenta/metabolism , Antigens, Protozoan/metabolism , Antibodies, Protozoan/metabolism , Erythrocytes/metabolism , Immunoglobulin M/metabolism
2.
PLoS Pathog ; 19(6): e1011442, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37307293

ABSTRACT

A signature remains elusive of naturally-acquired immunity against Plasmodium falciparum. We identified P. falciparum in a 14-month cohort of 239 people in Kenya, genotyped at immunogenic parasite targets expressed in the pre-erythrocytic (circumsporozoite protein, CSP) and blood (apical membrane antigen 1, AMA-1) stages, and classified into epitope type based on variants in the DV10, Th2R, and Th3R epitopes in CSP and the c1L region of AMA-1. Compared to asymptomatic index infections, symptomatic malaria was associated with reduced reinfection by parasites bearing homologous CSP-Th2R (adjusted hazard ratio [aHR]:0.63; 95% CI:0.45-0.89; p = 0.008) CSP-Th3R (aHR:0.71; 95% CI:0.52-0.97; p = 0.033), and AMA-1 c1L (aHR:0.63; 95% CI:0.43-0.94; p = 0.022) epitope types. The association of symptomatic malaria with reduced hazard of homologous reinfection was strongest for rare epitope types. Symptomatic malaria provides more durable protection against reinfection with parasites bearing homologous epitope types. The phenotype represents a legible molecular epidemiologic signature of naturally-acquired immunity by which to identify new antigen targets.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Malaria , Parasites , Animals , Plasmodium falciparum/metabolism , Reinfection , Protozoan Proteins/metabolism , Malaria/parasitology , Malaria, Falciparum/parasitology , Antigens, Protozoan , Epitopes/genetics , Antibodies, Protozoan/metabolism
3.
Malar J ; 21(1): 71, 2022 Mar 04.
Article in English | MEDLINE | ID: mdl-35246142

ABSTRACT

BACKGROUND: Plasmodium vivax is emerging as the dominant and prevalent species causing malaria in near-elimination settings outside of Africa. Hypnozoites, the dormant liver stage parasite of P. vivax, are undetectable to any currently available diagnostic test, yet are a major reservoir for transmission. Advances have been made to harness the naturally acquired immune response to identify recent exposure to P. vivax blood-stage parasites and, therefore, infer the presence of hypnozoites. This in-development diagnostic is currently able to detect infections within the last 9-months with 80% sensitivity and 80% specificity. Further work is required to optimize protein expression and protein constructs used for antibody detection. METHODS: The antibody response against the top performing predictor of recent infection, P. vivax reticulocyte binding protein 2b (PvRBP2b), was tested against multiple fragments of different sizes and from different expression systems. The IgG induced against the recombinant PvRBP2b fragments in P. vivax infected individuals was measured at the time of infection and in a year-long observational cohort; both conducted in Thailand. RESULTS: The antibody responses to some but not all different sized fragments of PvRBP2b protein are highly correlated with each other, significantly higher 1-week post-P. vivax infection, and show potential for use as predictors of recent P. vivax infection. CONCLUSIONS: To achieve P. vivax elimination goals, novel diagnostics are required to aid in detection of hidden parasite reservoirs. PvRBP2b was previously shown to be the top candidate for single-antigen classification of recent P. vivax exposure and here, it is concluded that several alternative recombinant PvRBP2b fragments can achieve equal sensitivity and specificity at predicting recent P. vivax exposure.


Subject(s)
Immunoglobulin G , Malaria, Vivax , Membrane Proteins , Plasmodium vivax , Protozoan Proteins , Antibodies, Protozoan/metabolism , Antibody Formation , Humans , Immunoglobulin G/metabolism , Malaria, Vivax/parasitology , Membrane Proteins/immunology , Peptide Fragments/immunology , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Reticulocytes/metabolism
4.
Nat Commun ; 13(1): 933, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177602

ABSTRACT

Understanding mechanisms of antibody synergy is important for vaccine design and antibody cocktail development. Examples of synergy between antibodies are well-documented, but the mechanisms underlying these relationships often remain poorly understood. The leading blood-stage malaria vaccine candidate, CyRPA, is essential for invasion of Plasmodium falciparum into human erythrocytes. Here we present a panel of anti-CyRPA monoclonal antibodies that strongly inhibit parasite growth in in vitro assays. Structural studies show that growth-inhibitory antibodies bind epitopes on a single face of CyRPA. We also show that pairs of non-competing inhibitory antibodies have strongly synergistic growth-inhibitory activity. These antibodies bind to neighbouring epitopes on CyRPA and form lateral, heterotypic interactions which slow antibody dissociation. We predict that such heterotypic interactions will be a feature of many immune responses. Immunogens which elicit such synergistic antibody mixtures could increase the potency of vaccine-elicited responses to provide robust and long-lived immunity against challenging disease targets.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , Antibodies, Protozoan/isolation & purification , Antibodies, Protozoan/metabolism , Antigens, Protozoan/genetics , Antigens, Protozoan/isolation & purification , Antigens, Protozoan/metabolism , Cell Line , Drosophila melanogaster , Epitopes/immunology , Humans , Immunogenicity, Vaccine , Malaria Vaccines/therapeutic use , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Protozoan Proteins/metabolism , Vaccine Development
5.
Elife ; 112022 01 13.
Article in English | MEDLINE | ID: mdl-35023832

ABSTRACT

Malaria is a global health burden, with Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) responsible for the majority of infections worldwide. Circumsporozoite protein (CSP) is the most abundant protein on the surface of Plasmodium sporozoites, and antibodies targeting the central repeat region of CSP can prevent parasite infection. Although much has been uncovered about the molecular basis of antibody recognition of the PfCSP repeats, data remains scarce for PvCSP. Here, we performed molecular dynamics simulations for peptides comprising the PvCSP repeats from strains VK210 and VK247 to reveal how the PvCSP central repeats are highly disordered, with minor propensities to adopt turn conformations. Next, we solved eight crystal structures to unveil the interactions of two inhibitory monoclonal antibodies (mAbs), 2F2 and 2E10.E9, with PvCSP repeats. Both antibodies can accommodate subtle sequence variances in the repeat motifs and recognize largely coiled peptide conformations that also contain isolated turns. Our structural studies uncover various degrees of Fab-Fab homotypic interactions upon recognition of the PvCSP central repeats by these two inhibitory mAbs, similar to potent mAbs against PfCSP. These findings augment our understanding of host-Plasmodium interactions and contribute molecular details of Pv inhibition by mAbs to unlock structure-based engineering of PvCSP-based vaccines.


Subject(s)
Antibodies, Protozoan/metabolism , Binding Sites, Antibody , Molecular Dynamics Simulation , Plasmodium vivax/immunology , Plasmodium vivax/metabolism , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Animals , Antibodies, Protozoan/chemistry , Cell Line , Crystallization , HEK293 Cells , Humans , Mice , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sporozoites/metabolism
6.
STAR Protoc ; 2(4): 100936, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34806047

ABSTRACT

This protocol describes the genomic phage (gPhage) display platform, a large-scale antigen and epitope mapping technique. We constructed a gPhage display peptide library of a eukaryotic organism, Trypanosoma cruzi (causative agent of Chagas disease), to map the antibody response landscape against the parasite. Here, we used an organism with a relatively large but intronless genome, although future applications could include other prevalent or (re)emerging infectious organisms carrying genomes with a limited number of introns. For complete details on the use and execution of this protocol, please refer to Teixeira et al. (2021).


Subject(s)
Cell Surface Display Techniques/methods , Genomic Library , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/metabolism , Genome, Protozoan/genetics , Trypanosoma cruzi/genetics
7.
Immunity ; 54(12): 2859-2876.e7, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34788599

ABSTRACT

Repeat antigens, such as the Plasmodium falciparum circumsporozoite protein (PfCSP), use both sequence degeneracy and structural diversity to evade the immune response. A few PfCSP-directed antibodies have been identified that are effective at preventing malaria infection, including CIS43, but how these repeat-targeting antibodies might be improved has been unclear. Here, we engineered a humanized mouse model in which B cells expressed inferred human germline CIS43 (iGL-CIS43) B cell receptors and used both vaccination and bioinformatic analysis to obtain variant CIS43 antibodies with improved protective capacity. One such antibody, iGL-CIS43.D3, was significantly more potent than the current best-in-class PfCSP-directed antibody. We found that vaccination with a junctional epitope peptide was more effective than full-length PfCSP at recruiting iGL-CIS43 B cells to germinal centers. Structure-function analysis revealed multiple somatic hypermutations that combinatorically improved protection. This mouse model can thus be used to understand vaccine immunogens and to develop highly potent anti-malarial antibodies.


Subject(s)
B-Lymphocyte Subsets/immunology , Epitopes/immunology , Malaria Vaccines/immunology , Malaria/immunology , Plasmodium falciparum/physiology , Protozoan Proteins/immunology , Vaccines, DNA/immunology , Adoptive Transfer , Animals , Antibodies, Protozoan/metabolism , Disease Models, Animal , Epitopes/genetics , Genetic Engineering , Humans , Immune Evasion , Immunogenicity, Vaccine , Mice , Mice, SCID , Protozoan Proteins/genetics , Structure-Activity Relationship , Vaccination
8.
Nat Commun ; 12(1): 5838, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34611164

ABSTRACT

Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) family members mediate receptor- and tissue-specific sequestration of infected erythrocytes (IEs) in malaria. Antibody responses are a central component of naturally acquired malaria immunity. PfEMP1-specific IgG likely protects by inhibiting IE sequestration and through IgG-Fc Receptor (FcγR) mediated phagocytosis and killing of antibody-opsonized IEs. The affinity of afucosylated IgG to FcγRIIIa is up to 40-fold higher than fucosylated IgG, resulting in enhanced antibody-dependent cellular cytotoxicity. Most IgG in plasma is fully fucosylated, but afucosylated IgG is elicited in response to enveloped viruses and to paternal alloantigens during pregnancy. Here we show that naturally acquired PfEMP1-specific IgG is strongly afucosylated in a stable and exposure-dependent manner, and efficiently induces FcγRIIIa-dependent natural killer (NK) cell degranulation. In contrast, immunization with a subunit PfEMP1 (VAR2CSA) vaccine results in fully fucosylated specific IgG. These results have implications for understanding protective natural- and vaccine-induced immunity to malaria.


Subject(s)
Antigens, Protozoan/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Antibodies, Protozoan/metabolism , Antigens, Protozoan/immunology , Female , Humans , Immunoglobulin G/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Pregnancy , Vaccination
10.
Nat Commun ; 12(1): 4226, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244481

ABSTRACT

RIFIN, a large family of Plasmodium variant surface antigens, plays a crucial role in malaria pathogenesis by mediating immune suppression through activation of inhibitory receptors such as LAIR1, and antibodies with LAIR1 inserts have been identified that bind infected erythrocytes through RIFIN. However, details of RIFIN-mediated LAIR1 recognition and receptor activation have been unclear. Here, we use negative-stain EM to define the architecture of LAIR1-inserted antibodies and determine crystal structures of RIFIN-variable 2 (V2) domain in complex with a LAIR1 domain. These structures reveal the LAIR1-binding region of RIFIN to be hydrophobic and membrane-distal, to exhibit extensive structural diversity, and to interact with RIFIN-V2 in a one-to-one fashion. Through structural and sequence analysis of various LAIR1 constructs, we identify essential elements of RIFIN-binding on LAIR1. Furthermore, a structure-derived LAIR1-binding sequence signature ascertained >20 LAIR1-binding RIFINs, including some from P. falciparum field strains and Plasmodium species infecting gorillas and chimpanzees.


Subject(s)
Antigens, Protozoan/ultrastructure , Malaria, Falciparum/immunology , Membrane Proteins/ultrastructure , Plasmodium falciparum/immunology , Protozoan Proteins/ultrastructure , Receptors, Immunologic/ultrastructure , Antibodies, Protozoan/genetics , Antibodies, Protozoan/metabolism , Antigenic Variation/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Crystallography, X-Ray , Humans , Malaria, Falciparum/parasitology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mutation , Plasmodium falciparum/metabolism , Protein Domains/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism
11.
PLoS One ; 16(7): e0254498, 2021.
Article in English | MEDLINE | ID: mdl-34252120

ABSTRACT

To screen for additional vaccine candidate antigens of Plasmodium pre-erythrocytic stages, fourteen P. falciparum proteins were selected based on expression in sporozoites or their role in establishment of hepatocyte infection. For preclinical evaluation of immunogenicity of these proteins in mice, chimeric P. berghei sporozoites were created that express the P. falciparum proteins in sporozoites as an additional copy gene under control of the uis4 gene promoter. All fourteen chimeric parasites produced sporozoites but sporozoites of eight lines failed to establish a liver infection, indicating a negative impact of these P. falciparum proteins on sporozoite infectivity. Immunogenicity of the other six proteins (SPELD, ETRAMP10.3, SIAP2, SPATR, HT, RPL3) was analyzed by immunization of inbred BALB/c and outbred CD-1 mice with viral-vectored (ChAd63 or ChAdOx1, MVA) vaccines, followed by challenge with chimeric sporozoites. Protective immunogenicity was determined by analyzing parasite liver load and prepatent period of blood stage infection after challenge. Of the six proteins only SPELD immunized mice showed partial protection. We discuss both the low protective immunogenicity of these proteins in the chimeric rodent malaria challenge model and the negative effect on P. berghei sporozoite infectivity of several P. falciparum proteins expressed in the chimeric sporozoites.


Subject(s)
Malaria, Falciparum/parasitology , Plasmodium falciparum/pathogenicity , Animals , Antibodies, Protozoan/immunology , Antibodies, Protozoan/metabolism , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Erythrocytes/metabolism , Female , Malaria Vaccines/therapeutic use , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Mice , Mice, Inbred BALB C , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Ribosomal Protein L3 , Sporozoites/pathogenicity
12.
J Immunol ; 206(8): 1817-1831, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33789984

ABSTRACT

Plasmodium falciparum merozoite surface protein (PfMSP)2 is a target of parasite-neutralizing Abs. Inclusion of recombinant PfMSP2 (rPfMSP2) as a component of a multivalent malaria vaccine is of interest, but presents challenges. Previously, we used the highly immunogenic PfMSP8 as a carrier to enhance production and/or immunogenicity of malaria vaccine targets. In this study, we exploited the benefits of rPfMSP8 as a carrier to optimize a rPfMSP2-based subunit vaccine. rPfMSP2 and chimeric rPfMSP2/8 vaccines produced in Escherichia coli were evaluated in comparative immunogenicity studies in inbred (CB6F1/J) and outbred (CD1) mice, varying the dose and adjuvant. Immunization of mice with both rPfMSP2-based vaccines elicited high-titer anti-PfMSP2 Abs that recognized the major allelic variants of PfMSP2. Vaccine-induced T cells recognized epitopes present in both PfMSP2 and the PfMSP8 carrier. Competition assays revealed differences in Ab specificities induced by the two rPfMSP2-based vaccines, with evidence of epitope masking by rPfMSP2-associated fibrils. In contrast to aluminum hydroxide (Alum) as adjuvant, formulation of rPfMSP2 vaccines with glucopyranosyl lipid adjuvant-stable emulsion, a synthetic TLR4 agonist, elicited Th1-associated cytokines, shifting production of Abs to cytophilic IgG subclasses. The rPfMSP2/8 + glucopyranosyl lipid adjuvant-stable emulsion formulation induced significantly higher Ab titers with superior durability and capacity to opsonize P. falciparum merozoites for phagocytosis. Immunization with a trivalent vaccine including PfMSP2/8, PfMSP1/8, and the P. falciparum 25 kDa sexual stage antigen fused to PfMSP8 (Pfs25/8) induced high levels of Abs specific for epitopes in each targeted domain, with no evidence of antigenic competition. These results are highly encouraging for the addition of rPfMSP2/8 as a component of an efficacious, multivalent, multistage malaria vaccine.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria/immunology , Merozoites/metabolism , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Th1 Cells/immunology , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Protozoan/metabolism , Antigens, Protozoan/genetics , Epitope Mapping , Female , Glucosides , Immunodominant Epitopes , Immunoglobulin G/metabolism , Lipid A , Malaria Vaccines/genetics , Male , Merozoites/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phagocytosis , Protozoan Proteins/genetics
13.
Elife ; 102021 03 02.
Article in English | MEDLINE | ID: mdl-33648633

ABSTRACT

Falciparum malaria is clinically heterogeneous and the relative contribution of parasite and host in shaping disease severity remains unclear. We explored the interaction between inflammation and parasite variant surface antigen (VSA) expression, asking whether this relationship underpins the variation observed in controlled human malaria infection (CHMI). We uncovered marked heterogeneity in the host response to blood challenge; some volunteers remained quiescent, others triggered interferon-stimulated inflammation and some showed transcriptional evidence of myeloid cell suppression. Significantly, only inflammatory volunteers experienced hallmark symptoms of malaria. When we tracked temporal changes in parasite VSA expression to ask whether variants associated with severe disease rapidly expand in naive hosts, we found no transcriptional evidence to support this hypothesis. These data indicate that parasite variants that dominate severe malaria do not have an intrinsic growth or survival advantage; instead, they presumably rely upon infection-induced changes in their within-host environment for selection.


Subject(s)
Antigenic Variation , Host-Pathogen Interactions/genetics , Malaria, Falciparum/immunology , Plasmodium falciparum/genetics , Adult , Animals , Anopheles/parasitology , Antibodies, Protozoan/genetics , Antibodies, Protozoan/metabolism , Antigens, Protozoan , Erythrocytes/immunology , Erythrocytes/parasitology , Female , Host-Pathogen Interactions/immunology , Humans , Inflammation , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Male , Plasmodium falciparum/pathogenicity , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
14.
mSphere ; 6(1)2021 01 06.
Article in English | MEDLINE | ID: mdl-33408232

ABSTRACT

Structure-guided vaccine design provides a route to elicit a focused immune response against the most functionally important regions of a pathogen surface. This can be achieved by identifying epitopes for neutralizing antibodies through structural methods and recapitulating these epitopes by grafting their core structural features onto smaller scaffolds. In this study, we conducted a modified version of this protocol. We focused on the PfEMP1 protein family found on the surfaces of erythrocytes infected with Plasmodium falciparum A subset of PfEMP1 proteins bind to endothelial protein C receptor (EPCR), and their expression correlates with development of the symptoms of severe malaria. Structural studies revealed that PfEMP1 molecules present a helix-kinked-helix motif that forms the core of the EPCR-binding site. Using Rosetta-based design, we successfully grafted this motif onto a three-helical bundle scaffold. We show that this synthetic binder interacts with EPCR with nanomolar affinity and adopts the expected structure. We also assessed its ability to bind to antibodies found in immunized animals and in humans from malaria-endemic regions. Finally, we tested the capacity of the synthetic binder to effectively elicit antibodies that prevent EPCR binding and analyzed the degree of cross-reactivity of these antibodies across a diverse repertoire of EPCR-binding PfEMP1 proteins. Despite our synthetic binder adopting the correct structure, we find that it is not as effective as the CIDRα domain on which it is based for inducing adhesion-inhibitory antibodies. This cautions against the rational design of focused immunogens that contain the core features of a ligand-binding site of a protein family, rather than those of a neutralizing antibody epitope.IMPORTANCE Vaccines train our immune systems to generate antibodies which recognize pathogens. Some of these antibodies are highly protective, preventing infection, while others are ineffective. Structure-guided rational approaches allow design of synthetic molecules which contain only the regions of a pathogen required to induce production of protective antibodies. On the surfaces of red blood cells infected by the malaria parasite Plasmodium falciparum are parasite molecules called PfEMP1 proteins. PfEMP1 proteins, which bind to human receptor EPCR, are linked to development of severe malaria. We have designed a synthetic protein on which we grafted the EPCR-binding surface of a PfEMP1 protein. We use this molecule to show which fraction of protective antibodies recognize the EPCR-binding surface and test its effectiveness as a vaccine immunogen.


Subject(s)
Antibodies, Protozoan/metabolism , Endothelial Protein C Receptor/metabolism , Proteins/chemical synthesis , Proteins/metabolism , Protozoan Proteins/agonists , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Motifs , Animals , Antibodies, Protozoan/immunology , Binding Sites , Cell Adhesion , Endothelial Protein C Receptor/immunology , Erythrocytes/parasitology , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Protein Binding , Proteins/chemistry , Proteins/immunology , Rats
15.
Emerg Microbes Infect ; 9(1): 2619-2621, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33215979

ABSTRACT

Toxoplasma gondii oocysts in the environment are a threat to humans and animals. This study is aimed to evaluate the prevalence of T. gondii in white spoonbills and isolate viable T. gondii from white spoonbills. In 28.6% (2/7) of white spoonbills, T. gondii antibodies were found in heart juice by the modified agglutination test (cut-off: 1:4). T. gondii DNA was detected in tissues of 42.9% (3/7) white spoonbills. One viable T. gondii strain, named TgSpoonbillCHn1, was isolated from the myocardium of a white spoonbill by bioassay in mice. DNA extracted from TgSpoonbillCHn1 tachyzoites was characterized by PCR-restriction fragment length polymorphism with ten markers and the virulence genes ROP5 and ROP18. The results revealed that it was ToxoDB#2 (Type III). The ROP18/ROP5 genotype combination predicts that this strain is avirulent for mice, which is supported by the infection experiments in mice. This is the first report of the isolation of viable T. gondii strain from white spoonbil. The prevalence of T. gondii in white spoonbills may be indicative of environmental contamination of oocysts. This report provides direct evidence of white spoonbill as an intermediate host of T. gondii.


Subject(s)
Antibodies, Protozoan/metabolism , Birds/virology , Toxoplasma/isolation & purification , Toxoplasmosis, Animal/diagnosis , Animals , China , DNA, Protozoan/genetics , Female , Heart/virology , Male , Mice , Species Specificity , Toxoplasma/genetics , Toxoplasma/immunology , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/immunology , Virulence
16.
Front Immunol ; 11: 1437, 2020.
Article in English | MEDLINE | ID: mdl-32733477

ABSTRACT

Babesiosis caused by Babesia species imposes an increasing threat to public-health and so far, there is no effective vaccine to prevent Babesia infections. Babesia surface antigen may participate in the invasion of erythrocytes. In our previous study, a surface antigen of B. microti merozoites, named as BmSP44 was identified as a dominant reactive antigen by protein microarray screening. To evaluate its potential applications in diagnosis and prevention of Babesiosis, the open reading frame encoding BmSP44 was cloned and the recombinant protein was expressed. In consistent with the protein microarray result, recombinant BmSP44 (rBmSP44) can be recognized by sera from B. microti infected mice. Immunofluorescence assays (IFA) confirmed that BmSP44 is a secreted protein and localized principally in the cytoplasm of the parasites. The parasitemia and Babesia gene copies were lower in mice administered rBmSP44 antisera compared with normal controls. Active immunization with rBmSP44 also afforded protection against B. microti infection. The concentrations of hemoglobin in rBmSP44 immunization group were higher than those in the control group. Importantly, vaccination of mice with rBmSP44 resulted in a Th1/Th2 mixed immune response with significantly elevated IL-10 and IFN-γ levels during the early stage of infection. Taken together, our results indicated that rBmSP44 can induce a protective immune response against Babesia infection. Thus, BmSP44 can be used as both a diagnosis marker and a vaccine candidate.


Subject(s)
Antibodies, Protozoan/metabolism , Antigens, Protozoan/metabolism , Babesia microti/physiology , Babesiosis/parasitology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Babesiosis/immunology , Cloning, Molecular , Disease Models, Animal , Disease Resistance , Female , Hemoglobins/metabolism , Humans , Immunity, Cellular , Merozoites , Mice , Mice, Inbred BALB C , Protein Array Analysis , Vaccination
17.
Sci Rep ; 10(1): 12102, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32694844

ABSTRACT

Efficient health-care for pregnant women require accurate data on the prevalence of toxoplasmosis in pregnancy at global, regional, and country levels. In this systematic review with meta- and modelling-analysis, we searched PubMed, EMBASE, Web of Knowledge, Global Index Medicus, and Africa Journal Online to identify studies that reported enough data to compute the immunoglobulins (Ig) M or G seroprevalence estimates of Toxoplasma gondii in pregnant women up to December 31st, 2018, without any language restriction. The global and regional estimates were done using a random-effects meta-analysis. We included 250 studies with 723,655 pregnant women. The global IgM seroprevalence was 1.9% (95%CI: 1.7-2.3). At the regional level, Eastern Mediterranean had the highest IgM seroprevalence (4.1%, 95%CI: 2.8-5.5) and The Americas, the lowest (1.1%, 0.8-1.4), with a statistically significant difference between WHO regions (p < 0.0001). The global IgG seroprevalence was 32.9% (95%CI: 29.4-36.4). Among WHO regions, The Americas had the highest prevalence (45.2%, 95%CI: 33.4-53.4) and Western Pacific the lowest (11.2%, 7.8-15.1), with a statistically significant difference between regions (p < 0.0001). This study presents a high toxoplasma seropositivity in pregnant women at global, regional and country levels, with a consequential high risk of maternal and congenital toxoplasmosis.


Subject(s)
Immunoglobulin M/metabolism , Pregnancy Complications, Parasitic/epidemiology , Toxoplasma/immunology , Toxoplasmosis/epidemiology , Americas/epidemiology , Antibodies, Protozoan/metabolism , Female , Global Health , Humans , Mediterranean Region/epidemiology , Pacific States/epidemiology , Pregnancy , Pregnancy Complications, Parasitic/immunology , Seroepidemiologic Studies , Toxoplasmosis/immunology
18.
Nat Immunol ; 21(7): 790-801, 2020 07.
Article in English | MEDLINE | ID: mdl-32424361

ABSTRACT

Plasmodium parasite-specific antibodies are critical for protection against malaria, yet the development of long-lived and effective humoral immunity against Plasmodium takes many years and multiple rounds of infection and cure. Here, we report that the rapid development of short-lived plasmablasts during experimental malaria unexpectedly hindered parasite control by impeding germinal center responses. Metabolic hyperactivity of plasmablasts resulted in nutrient deprivation of the germinal center reaction, limiting the generation of memory B cell and long-lived plasma cell responses. Therapeutic administration of a single amino acid to experimentally infected mice was sufficient to overcome the metabolic constraints imposed by plasmablasts and enhanced parasite clearance and the formation of protective humoral immune memory responses. Thus, our studies not only challenge the current model describing the role and function of blood-stage Plasmodium-induced plasmablasts but they also reveal new targets and strategies to improve anti-Plasmodium humoral immunity.


Subject(s)
Immunity, Humoral , Malaria/immunology , Plasma Cells/metabolism , Plasmodium falciparum/immunology , Adolescent , Adult , Amino Acids/administration & dosage , Amino Acids/metabolism , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antibodies, Protozoan/metabolism , Antimalarials/administration & dosage , DNA, Protozoan/isolation & purification , Disease Models, Animal , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Host-Parasite Interactions/immunology , Humans , Malaria/blood , Malaria/drug therapy , Malaria/parasitology , Mice , Mice, Transgenic , Middle Aged , Nutrients/metabolism , Plasma Cells/immunology , Plasma Cells/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Proof of Concept Study , Young Adult
19.
Poult Sci ; 99(1): 374-384, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32416822

ABSTRACT

Research has shown that methionine+ cysteine (M+C) requirements may be higher when chickens are infected with Eimeria app. In a 4 × 2 factorial design, broilers (11 to 21 D) were fed one of 4 corn-soybean meal-based diets containing either 0.6, 0.8, 0.9, or 1.0% standardized ileal digestible (SID) M+C; on day 14, broilers from each diet were gavaged with either phosphate-buffered saline (PBS) or a commercial coccidiosis vaccine (at 100 × vaccine dose) which provide a mixture of live Eimeria acervulina, Eimeria maxima, and Eimeria tenella oocysts. Growth performance was recorded from day 11 to 21. Plasma and intestinal luminal samples were collected on days 14 and 21. Intestine lesion scores and fecal oocyst counts were conducted on day 21. Regardless of dietary SID M+C levels, compared to PBS gavaged broilers, the Eimeria-challenged broilers had (1) decreased (P < 0.05) body weight gain (BWG), feed intake (FI), and gain-to-feed ratio (G:F); (2) increased (P < 0.05) intestinal lesion scores and fecal oocyst counts; (3) increased (P < 0.05) plasma anti-Eimeria IgG, and intestinal luminal total IgA and anti-Eimeria IgA concentrations; and (4) increased (P < 0.05) levels of duodenum luminal gamma interferon (IFN-γ) and interleukin-10 (IL-10), as well as jejunum and cecum luminal IFN-γ concentrations. Regardless of Eimeria challenge, when compared to 0.6% SID M+C, broilers fed ≥0.8% SID M+C had (1) increased (P < 0.05) BWG, FI, and G:F and (2) increased (P < 0.05) levels of jejunum luminal total IgA. After Eimeria challenge, broilers fed 0.8% SID M+C had increased (P < 0.05) levels of jejunum luminal anti-Eimeria IgA compared to broilers fed diets containing 0.6 and 1.0% SID M+C. Collectively, in 11- to 21-D broilers, the growth suppression caused by Eimeria infection could not be mitigated by further increasing dietary M+C alone ≥0.8%. Further research should investigate interactions between dietary M+C and other nutrients for support of immune function and growth in pathogen-challenged broilers.


Subject(s)
Chickens/immunology , Cysteine/pharmacology , Methionine/pharmacology , Poultry Diseases/parasitology , Animal Feed/analysis , Animals , Antibodies, Protozoan/metabolism , Chickens/growth & development , Coccidiosis/prevention & control , Coccidiosis/veterinary , Cysteine/administration & dosage , Diet/veterinary , Eimeria/physiology , Intestines/immunology , Male , Methionine/administration & dosage , Oocysts , Poultry Diseases/immunology
20.
Front Immunol ; 11: 412, 2020.
Article in English | MEDLINE | ID: mdl-32210975

ABSTRACT

P27A is a novel synthetic malaria vaccine candidate derived from the blood stage Plasmodium falciparum protein Trophozoite Exported Protein 1 (TEX1/PFF0165c). In phase 1a/1b clinical trials in malaria unexposed adults in Switzerland and in malaria pre-exposed adults in Tanzania, P27A formulated with Alhydrogel and GLA-SE adjuvants induced antigen-specific antibodies and T-cell activity. The GLA-SE adjuvant induced significantly stronger humoral responses than the Alhydrogel adjuvant. Groups of pre-exposed and unexposed subjects received identical vaccine formulations, which supported the comparison of the cellular and humoral response to P27A in terms of fine specificity and affinity for populations and adjuvants. Globally, fine specificity of the T and B cell responses exhibited preferred recognized sequences and did not highlight major differences between adjuvants or populations. Affinity of anti-P27A antibodies was around 10-8 M in all groups. Pre-exposed volunteers presented anti-P27A with higher affinity than unexposed volunteers. Increasing the dose of GLA-SE from 2.5 to 5 µg in pre-exposed volunteers improved anti-P27A affinity and decreased the number of recognized epitopes. These results indicate a higher maturation of the humoral response in pre-exposed volunteers, particularly when immunized with P27A formulated with 5 µg GLA-SE.


Subject(s)
Antigens, Protozoan/immunology , Epitope Mapping/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Peptides/immunology , Protozoan Proteins/immunology , Adjuvants, Immunologic , Adult , Antibodies, Protozoan/metabolism , Antibody Affinity , Antigens, Protozoan/genetics , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte/genetics , Humans , Life Cycle Stages , Lymphocyte Activation , Peptides/genetics , Plasmodium falciparum , Protozoan Proteins/genetics , Switzerland , Tanzania , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...