Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Sci Rep ; 12(1): 3040, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197516

ABSTRACT

The pathogenesis of malaria is associated with blood-stage infection and there is strong evidence that antibodies specific to parasite blood-stage antigens can control parasitemia. This provides a strong rational for applying blood-stage antigen components in a multivalent vaccine, as the induced antibodies in combination can enhance protection. The Plasmodium falciparum rhoptry-associated membrane antigen (PfRAMA) is a promising vaccine target, due to its fundamental role in merozoite invasion and low level of polymorphism. Polyclonal antibodies against PfRAMA are able to inhibit P. falciparum growth and interact synergistically when combined with antibodies against P. falciparum reticulocyte-binding protein 5 (PfRh5) or cysteine-rich protective antigen (PfCyRPA). In this study, we identified a novel PfRAMA-specific mAb with neutralizing activity, which in combination with PfRh5- or PfCyRPA-specific mAbs potentiated the neutralizing effect. By applying phage display technology, we mapped the protective epitope to be in the C-terminal region of PfRAMA. Our results confirmed previous finding of synergy between PfRAMA-, PfRh5- and PfCyRPA-specific antibodies, thereby paving the way of testing these antigens (or fragments of these antigens) in combination to improve the efficacy of blood-stage malaria vaccines. The results emphasize the importance of directing antibody responses towards protective epitopes, as the majority of anti-PfRAMA mAbs were unable to inhibit merozoite invasion of erythrocytes.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/isolation & purification , Antibodies, Protozoan/chemistry , Antigens, Protozoan/immunology , Carrier Proteins/immunology , Cell Line , Drug Synergism , Epitopes/chemistry , Epitopes/immunology , Humans , Malaria Vaccines/chemistry , Malaria, Falciparum/prevention & control , Merozoites/immunology , Mice , Protein Binding , Protozoan Proteins/biosynthesis , Protozoan Proteins/chemistry , Protozoan Proteins/isolation & purification
2.
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
3.
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
4.
PLoS Pathog ; 17(9): e1009920, 2021 09.
Article in English | MEDLINE | ID: mdl-34547035

ABSTRACT

RTX leukotoxins are a diverse family of prokaryotic virulence factors that are secreted by the type 1 secretion system (T1SS) and target leukocytes to subvert host defenses. T1SS substrates all contain a C-terminal RTX domain that mediates recruitment to the T1SS and drives secretion via a Brownian ratchet mechanism. Neutralizing antibodies against the Bordetella pertussis adenylate cyclase toxin, an RTX leukotoxin essential for B. pertussis colonization, have been shown to target the RTX domain and prevent binding to the αMß2 integrin receptor. Knowledge of the mechanisms by which antibodies bind and neutralize RTX leukotoxins is required to inform structure-based design of bacterial vaccines, however, no structural data are available for antibody binding to any T1SS substrate. Here, we determine the crystal structure of an engineered RTX domain fragment containing the αMß2-binding site bound to two neutralizing antibodies. Notably, the receptor-blocking antibodies bind to the linker regions of RTX blocks I-III, suggesting they are key neutralization-sensitive sites within the RTX domain and are likely involved in binding the αMß2 receptor. As the engineered RTX fragment contained these key epitopes, we assessed its immunogenicity in mice and showed that it elicits similar neutralizing antibody titers to the full RTX domain. The results from these studies will support the development of bacterial vaccines targeting RTX leukotoxins, as well as next-generation B. pertussis vaccines.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/chemistry , Pertussis Vaccine , Virulence Factors, Bordetella/chemistry , Adenylate Cyclase Toxin/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Bordetella pertussis , Mice , Protein Domains/immunology , Virulence Factors, Bordetella/immunology , Whooping Cough/immunology , Whooping Cough/prevention & control
5.
Am J Trop Med Hyg ; 104(1): 229-232, 2021 01.
Article in English | MEDLINE | ID: mdl-33078702

ABSTRACT

Cryptosporidiosis is common in early childhood, and both diarrheal and subclinical infections are associated with adverse developmental outcomes. Improved therapeutic medications may help reduce the burden of cryptosporidial diarrhea; however, an effective vaccine would be better able to prevent the detrimental impact of both diarrheal and subclinical disease. A more complete understanding of naturally occurring immunity may further inform strategies to develop an effective vaccine. In this prospective cohort study of Bangladeshi children, greater fecal IgA at 12 months, but not plasma IgG, directed against two sporozoite-expressed, immunodominant and vaccine candidate antigens was associated with delayed time to subsequent cryptosporidiosis to 3 years of life. These findings extend prior work and further support the role of mucosal antibody responses in naturally developing protective immunity to Cryptosporidium.


Subject(s)
Antibodies, Protozoan/chemistry , Antigens, Protozoan/immunology , Cryptosporidium/immunology , Feces/parasitology , Immunoglobulin A/immunology , Antibodies, Protozoan/blood , Bangladesh/epidemiology , Child, Preschool , Cryptosporidiosis , Cryptosporidium/metabolism , Female , Humans , Immunoglobulin G/blood , Infant , Male
6.
PLoS Pathog ; 16(3): e1008373, 2020 03.
Article in English | MEDLINE | ID: mdl-32150583

ABSTRACT

Lasting protection has long been a goal for malaria vaccines. The major surface antigen on Plasmodium falciparum sporozoites, the circumsporozoite protein (PfCSP), has been an attractive target for vaccine development and most protective antibodies studied to date interact with the central NANP repeat region of PfCSP. However, it remains unclear what structural and functional characteristics correlate with better protection by one antibody over another. Binding to the junctional region between the N-terminal domain and central NANP repeats has been proposed to result in superior protection: this region initiates with the only NPDP sequence followed immediately by NANP. Here, we isolated antibodies in Kymab mice immunized with full-length recombinant PfCSP and two protective antibodies were selected for further study with reactivity against the junctional region. X-ray and EM structures of two monoclonal antibodies, mAb667 and mAb668, shed light on their differential affinity and specificity for the junctional region. Importantly, these antibodies also bind to the NANP repeat region with equal or better affinity. A comparison with an NANP-only binding antibody (mAb317) revealed roughly similar but statistically distinct levels of protection against sporozoite challenge in mouse liver burden models, suggesting that junctional antibody protection might relate to the ability to also cross-react with the NANP repeat region. Our findings indicate that additional efforts are necessary to isolate a true junctional antibody with no or much reduced affinity to the NANP region to elucidate the role of the junctional epitope in protection.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Protozoan/chemistry , Binding Sites, Antibody , Epitopes/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Protozoan/immunology , Epitopes/immunology , Female , Male , Mice , Mice, Transgenic , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Structure-Activity Relationship
7.
Nat Commun ; 10(1): 4328, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31551421

ABSTRACT

Transmission-blocking vaccines have the potential to be key contributors to malaria elimination. Such vaccines elicit antibodies that inhibit parasites during their development in Anopheles mosquitoes, thus breaking the cycle of transmission. To date, characterization of humoral responses to Plasmodium falciparum transmission-blocking vaccine candidate Pfs25 has largely been conducted in pre-clinical models. Here, we present molecular analyses of human antibody responses generated in a clinical trial evaluating Pfs25 vaccination. From a collection of monoclonal antibodies with transmission-blocking activity, we identify the most potent transmission-blocking antibody yet described against Pfs25; 2544. The interactions of 2544 and three other antibodies with Pfs25 are analyzed by crystallography to understand structural requirements for elicitation of human transmission-blocking responses. Our analyses provide insights into Pfs25 immunogenicity and epitope potency, and detail an affinity maturation pathway for a potent transmission-blocking antibody in humans. Our findings can be employed to guide the design of improved malaria transmission-blocking vaccines.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Antibodies, Protozoan/chemistry , Antibody Formation , Binding Sites, Antibody , Crystallography, X-Ray , Humans , Malaria, Falciparum/transmission , Protozoan Proteins/chemistry
8.
Nat Microbiol ; 4(9): 1497-1507, 2019 09.
Article in English | MEDLINE | ID: mdl-31133755

ABSTRACT

The most widespread form of malaria is caused by Plasmodium vivax. To replicate, this parasite must invade immature red blood cells through a process requiring interaction of the P. vivax Duffy binding protein (PvDBP) with its human receptor, the Duffy antigen receptor for chemokines. Naturally acquired antibodies that inhibit this interaction associate with clinical immunity, suggesting PvDBP as a leading candidate for inclusion in a vaccine to prevent malaria due to P. vivax. Here, we isolated a panel of monoclonal antibodies from human volunteers immunized in a clinical vaccine trial of PvDBP. We screened their ability to prevent PvDBP from binding to the Duffy antigen receptor for chemokines, and their capacity to block red blood cell invasion by a transgenic Plasmodium knowlesi parasite genetically modified to express PvDBP and to prevent reticulocyte invasion by multiple clinical isolates of P. vivax. This identified a broadly neutralizing human monoclonal antibody that inhibited invasion of all tested strains of P. vivax. Finally, we determined the structure of a complex of this antibody bound to PvDBP, indicating the molecular basis for inhibition. These findings will guide future vaccine design strategies and open up possibilities for testing the prophylactic use of such an antibody.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Receptors, Cell Surface/immunology , Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Crystallography, X-Ray , Duffy Blood-Group System/metabolism , Epitopes, B-Lymphocyte , Erythrocytes/parasitology , Genetic Variation , Humans , Immunoglobulin Fab Fragments , Malaria Vaccines/administration & dosage , Malaria, Vivax/parasitology , Plasmodium knowlesi/genetics , Plasmodium knowlesi/growth & development , Plasmodium knowlesi/immunology , Plasmodium vivax/genetics , Plasmodium vivax/growth & development , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Reticulocytes/parasitology
9.
Int J Mol Sci ; 20(8)2019 Apr 12.
Article in English | MEDLINE | ID: mdl-31013713

ABSTRACT

Two Leishmania infantum mimotopes (B10 and C01) identified by phage display showed to be antigenic and immunogenic for visceral (VL) and tegumentary (TL) leishmaniasis; however, their biological targets in the parasites have not been identified. The aim of the present study was to investigate the native antigens expressing both mimotopes, and to use them in distinct immunological assays. For this, a subtractive phage display technology was used, where a combinatorial library of single-chain variable fragments (scFv) was employed and the most reactive monoclonal antibodies for each target were captured, being the target antigens identified by mass spectrometry. Results in immunoblotting and immunoprecipitation assays showed that both monoclonal scFvs antibodies identified the ß-tubulin protein as the target antigen in L. infantum. To validate these findings, the recombinant protein was cloned, purified and tested for the serodiagnosis of human leishmaniasis, and its immunogenicity was evaluated in PBMC derived from healthy subjects and treated or untreated VL patients. Results showed high diagnostic efficacy, as well as the development of a specific Th1 immune response in the cell cultures, since higher IFN-γ and lower IL-10 production was found.


Subject(s)
Leishmania infantum/genetics , Leishmania infantum/metabolism , Leishmaniasis, Visceral/parasitology , Tubulin/metabolism , Amino Acid Sequence , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/immunology , Cell Surface Display Techniques , Cytokines/metabolism , Humans , Leishmania infantum/drug effects , Leishmania infantum/immunology , Leishmaniasis, Visceral/diagnosis , Leishmaniasis, Visceral/drug therapy , Models, Molecular , Protein Conformation , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Theranostic Nanomedicine , Tubulin/genetics , Tubulin/immunology
10.
Sci Adv ; 4(10): eaau8529, 2018 10.
Article in English | MEDLINE | ID: mdl-30324137

ABSTRACT

The circumsporozoite protein (CSP) on the surface of Plasmodium falciparum sporozoites is important for parasite development, motility, and host hepatocyte invasion. However, intrinsic disorder of the NANP repeat sequence in the central region of CSP has hindered its structural and functional characterization. Here, the cryo-electron microscopy structure at ~3.4-Å resolution of a recombinant shortened CSP construct with the variable domains (Fabs) of a highly protective monoclonal antibody reveals an extended spiral conformation of the central NANP repeat region surrounded by antibodies. This unusual structure appears to be stabilized and/or induced by interaction with an antibody where contacts between adjacent Fabs are somatically mutated and enhance the interaction. This maturation in non-antigen contact residues may be an effective mechanism for antibodies to target tandem repeat sequences and provide novel insights into malaria vaccine design.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Cryoelectron Microscopy/methods , Immunoglobulin Fab Fragments/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Sporozoites/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/genetics , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Immunoglobulin Fab Fragments/genetics , Malaria/immunology , Malaria/prevention & control , Mice , Mutation , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Conformation , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Sequence Homology , Sporozoites/metabolism
11.
Nat Commun ; 9(1): 4458, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367064

ABSTRACT

Interventions that can block the transmission of malaria-causing Plasmodium falciparum (Pf) between the human host and Anopheles vector have the potential to reduce the incidence of malaria. Pfs48/45 is a gametocyte surface protein critical for parasite development and transmission, and its targeting by monoclonal antibody (mAb) 85RF45.1 leads to the potent reduction of parasite transmission. Here, we reveal how the Pfs48/45 6C domain adopts a (SAG1)-related-sequence (SRS) fold. We structurally delineate potent epitope I and show how mAb 85RF45.1 recognizes an electronegative surface with nanomolar affinity. Analysis of Pfs48/45 sequences reveals that polymorphisms are rare for residues involved at the binding interface. Humanization of rat-derived mAb 85RF45.1 conserved the mode of recognition and activity of the parental antibody, while also improving its thermostability. Our work has implications for the development of transmission-blocking interventions, both through improving vaccine designs and the testing of passive delivery of mAbs in humans.


Subject(s)
Antigens, Protozoan/chemistry , Epitopes/chemistry , Malaria, Falciparum/transmission , Membrane Glycoproteins/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/immunology , Antigen-Antibody Reactions , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Epitopes/immunology , Humans , Malaria Vaccines , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Models, Molecular , Protein Domains , Protein Folding , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Rats
12.
Parasite Immunol ; 40(11): e12585, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30132929

ABSTRACT

Trypanosoma cruzi 24 (Tc24) is a recently described B-cell superantigen (BC-SAg) expressed by all developmental stages of T. cruzi, the causative agent of Chagas disease. BC-SAgs are immunoevasins that interfere with the catalytic response available to a subset of natural antibodies comprising the preimmune (innate) repertoire. Electrophilic modifications of BC-SAgs facilitate the formation of highly energetic covalent reactions favouring B-cell differentiation instead of B-cell downregulation. Therefore, the aim of this study was to convert the inhibitory signals delivered to B-cells with specificity for Tc24 into activating signals after conjugating electrophilic phosphonate groups to recombinant Tc24 (eTc24). Covalent immunization with eTc24 increased the binding affinity between eTc24 and naturally nucleophilic immunoglobulins with specificity for this BC-SAg. Flow cytometric analyses demonstrated that eTc24 but not Tc24 or other electrophilically modified control proteins bound Tc24-specific IgM+ B-cells covalently. In addition, immunization of mice with eTc24 adjuvanted with ISA720 induced the production of catalytic responses specific for Tc24 compared to the abrogation of this response in mice immunized with Tc24/ISA720. eTc24-immunized mice also produced IgMs that bound recombinant Tc24 compared to the binding observed for IgMs purified from non eTc24-immunized controls. These data suggest that eTc24 immunization overrides the immunosuppressive properties of this BC-SAg.


Subject(s)
Antibodies, Catalytic/immunology , Antibodies, Protozoan/immunology , Chagas Disease/immunology , Protozoan Proteins/immunology , Trypanosoma cruzi/immunology , Animals , Antibodies, Protozoan/chemistry , Antibody Formation , B-Lymphocytes/immunology , Chagas Disease/diagnosis , Chagas Disease/parasitology , Female , Humans , Immunization , Immunoglobulin M/immunology , Mice , Mice, Inbred BALB C , Protozoan Proteins/chemistry , Trypanosoma cruzi/chemistry , Trypanosoma cruzi/genetics , Vaccination
13.
J Immunol Methods ; 460: 87-92, 2018 09.
Article in English | MEDLINE | ID: mdl-30056943

ABSTRACT

Trypanosoma evansi, a haemoflagellated protozoan parasite, is responsible for chronic as well as the acute debilitating disease called surra in a wide range of herbivores and carnivores including domestic and wild animals. Since the parasite is having wide host range, there is a need for diagnostic test which can detect the T. evansi specific antibody in different species of animals for generating sero-surveillance data. In the present study we developed and evaluated competitive enzyme immunoassay using monoclonal antibodies (MAbs) raised against recombinant variable surface glycoprotein (rVSG) of T. evansi. The immunoreactivity of the developed MAbs (IgG3-subtype) was evaluated by immunoblot as well as ELISA and subsequently used in the development and standardization of competitive ELISA (C-ELISA). Further, the serological data generated from the C-ELISA using reference samples constituting true positive or surely infected (35), true negative (45), sero-positive (225) and sero-negative (215) samples and was analyzed statistically. The true positivity/negativity was determined by thin blood smear examination and diagnostic PCR assay, While, seropositivity/seronegativity of the reference samples was determined through standard reference tests. The data showed the diagnostic sensitivity of 92.6% and specificity of 96.4% with Cohen's kappa value of 0.88. In order to determine the utility of C-ELISA in detecting T. evansi antibodies in different species of animals, the assay was further evaluated with 1361 field sera sample comprising bovine, horse, donkey and camel. Since the C-ELISA described herein has showed high sensitivity and specificity, this single test can be explored in the sero-surveillance of T. evansi in a wide range of animals.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Protozoan/chemistry , Antigens, Protozoan , Protozoan Proteins , Trypanosoma , Trypanosomiasis , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Buffaloes , Camelus , Cattle , Enzyme-Linked Immunosorbent Assay , Equidae , Horses , Immunoblotting , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Trypanosoma/genetics , Trypanosoma/immunology , Trypanosomiasis/diagnosis , Trypanosomiasis/immunology , Trypanosomiasis/veterinary
14.
Protein Expr Purif ; 152: 122-130, 2018 12.
Article in English | MEDLINE | ID: mdl-30059744

ABSTRACT

Plants as a platform for recombinant protein expression are now economically comparable to well-established systems, such as microbes and mammalian cells, thanks to advantages such as scalability and product safety. However, downstream processing accounts for the majority of the final product costs because plant extracts contain large quantities of host cell proteins (HCPs) that must be removed using elaborate purification strategies. Heat precipitation in planta (blanching) can remove ∼80% of HCPs and thus simplify further purification steps, but this is only possible if the target protein is thermostable. Here we describe a combination of blanching and chromatography to purify the thermostable transmission-blocking malaria vaccine candidate FQS, which was transiently expressed in Nicotiana benthamiana leaves. If the blanching temperature exceeded a critical threshold of ∼75 °C, FQS was no longer recognized by the malaria transmission-blocking monoclonal antibody 4B7. A design-of-experiments approach revealed that reducing the blanching temperature from 80 °C to 70 °C restored antibody binding while still precipitating most HCPs. We also found that blanching inhibited the degradation of FQS in plant extracts, probably due to the thermal inactivation of proteases. We screened hydrophobic interaction chromatography materials using miniature columns and a liquid-handling station. Octyl Sepharose achieved the highest FQS purity during the primary capture step and led to a final purity of ∼72% with 60% recovery via step elution. We found that 30-75% FQS was lost during ultrafiltration/diafiltration, giving a final yield of 9 mg kg-1 plant material after purification based on an initial yield of ∼49 mg kg-1 biomass after blanching.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Protozoan/chemistry , Malaria Vaccines/isolation & purification , Nicotiana/genetics , Plant Proteins/isolation & purification , Protozoan Proteins/isolation & purification , Antibodies, Monoclonal/metabolism , Antibodies, Protozoan/metabolism , Chromatography, Affinity/methods , Cloning, Molecular , Factor Analysis, Statistical , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hot Temperature , Malaria Vaccines/biosynthesis , Malaria Vaccines/genetics , Plant Leaves/genetics , Plant Leaves/metabolism , Plant Proteins/chemistry , Plants, Genetically Modified , Protein Binding , Protein Denaturation , Protozoan Proteins/biosynthesis , Protozoan Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sepharose/analogs & derivatives , Nicotiana/chemistry , Nicotiana/metabolism , Ultrafiltration/methods
15.
Science ; 360(6395): 1358-1362, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29880723

ABSTRACT

Affinity maturation selects B cells expressing somatically mutated antibody variants with improved antigen-binding properties to protect from invading pathogens. We determined the molecular mechanism underlying the clonal selection and affinity maturation of human B cells expressing protective antibodies against the circumsporozoite protein of the malaria parasite Plasmodium falciparum (PfCSP). We show in molecular detail that the repetitive nature of PfCSP facilitates direct homotypic interactions between two PfCSP repeat-bound monoclonal antibodies, thereby improving antigen affinity and B cell activation. These data provide a mechanistic explanation for the strong selection of somatic mutations that mediate homotypic antibody interactions after repeated parasite exposure in humans. Our findings demonstrate a different mode of antigen-mediated affinity maturation to improve antibody responses to PfCSP and presumably other repetitive antigens.


Subject(s)
Antibodies, Protozoan/immunology , Antibody Affinity/immunology , Antibody Formation/immunology , Antigens, Protozoan/immunology , B-Lymphocytes/immunology , Epitopes, B-Lymphocyte/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/genetics , Antibody Affinity/genetics , Antibody Formation/genetics , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Humans , Lymphocyte Activation , Mutation , Protozoan Proteins/genetics , Repetitive Sequences, Nucleic Acid/genetics , Repetitive Sequences, Nucleic Acid/immunology , Selection, Genetic
16.
Monoclon Antib Immunodiagn Immunother ; 37(2): 87-90, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29634395

ABSTRACT

Malaria is a life-threatening infectious disease and continues to be a major public health crisis in many parts of the tropical world. Plasmodium falciparum is responsible for the majority of mortality and morbidity associated with malaria. During the intraerythrocytic cycle, P. falciparum releases three proteins with high histidine content as follows: histidine-rich protein 1 (HRP1), histidine-rich protein 2 (HRP2), and histidine-rich protein 3 (HRP3). Currently, most of the diagnostic tests of P. falciparum infection target HRP2, and a number of monoclonal antibodies (mAbs) against HRP2 have been developed for use in HRP2 detection and quantification. When parasites have HRP2 deletions, the detection of HRP3 could augment the sensitivity of the detection system. The combination of both HRP2 and HRP3 mAbs in the detection system will enhance the test sensitivity. In the HRP quantitative enzyme-linked immunosorbent assay (ELISA), both HRP2 and HRP3 contribute to the result, but the relative contribution of HRP2 and HRP3 was unable to investigate, because of the nonavailability of HRP3 specific antibody ELISA. Hence an ELISA test system based on HRP3 is also essential for detection and quantification. There is not much documented in the literature on HRP3 antigen and HRP3 specific mAbs and polyclonal antibodies (pAbs). In the present study, recombinant HRP3 was expressed in Escherichia coli and purified with Ni-NTA agarose column. The purified rHRP3 was used for the generation and characterization of monoclonal and pAbs. The purification of monoclonal and pAbs was done using a mixed-mode chromatography sorbent, phenylpropylamine HyperCel™. With the purified antibodies, a sandwich ELISA was developed. The sandwich ELISA method was explored to detect and quantify HRP3 of P. falciparum in the spent medium. The generated mAbs could be potentially used for the detection and quantification of P. falciparum HRP3.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Protozoan/biosynthesis , Antibody Specificity , Antigens, Protozoan/administration & dosage , Enzyme-Linked Immunosorbent Assay/methods , Plasmodium falciparum/immunology , Protozoan Proteins/administration & dosage , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/isolation & purification , Antigens, Protozoan/analysis , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Cell Fusion , Cells, Cultured , Cloning, Molecular , Culture Media, Conditioned/chemistry , Erythrocytes/parasitology , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Hybridomas/chemistry , Hybridomas/immunology , Immunization, Secondary , Mice , Mice, Inbred BALB C , Protozoan Proteins/analysis , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spleen/cytology , Spleen/immunology
17.
Am J Trop Med Hyg ; 99(1): 51-56, 2018 07.
Article in English | MEDLINE | ID: mdl-29637884

ABSTRACT

Babesia is a tick-borne intraerythrocytic parasite that is clinically and diagnostically similar to malaria parasite, conferring risk of misdiagnosis in areas where both parasites are endemic. Data on Babesia in humans in Africa are lacking, despite evidence that it is present in regional animal populations. Samples that were collected in November 2014 to July 2015 in Kilosa district, Tanzania, were evaluated for evidence of malaria and Babesia infection. Clinical data and laboratory samples (i.e., hemoglobin, rapid diagnostic testing [RDT] for malaria, peripheral blood smear, and dried blood spots) from a routine survey were available for analysis. Dried blood spots were tested using an investigational enzyme linked immunosorbent assay (ELISA) against Babesia microti. A total of 1,030 children aged 1 month to < 5 years were evaluated; 186 (18.1%) were malaria RDT positive, 180 (96.8%) of whom had peripheral smears reviewed; 70/180 (38.9%) were smear positive for parasites. The median (inter quartile range) and range of B. microti ELISA signal to cutoff (S/C) ratio was 0.10 (0.06-0.15) and 0.01-1.65, respectively; the S/C ratios were significantly higher in subjects ≥ 1 year as compared with those < 1 year old (P < 0.001). There was also a statistically significant association between a positive RDT for malaria and the Babesia S/C (median 0.09 versus 0.13 in RDT negative versus RDT positive, respectively; P < 0.001). The highest S/C ratios were disproportionately clustered in a few hamlets. The findings suggest that Babesia may be present in Kilosa district, Tanzania. However, serological cross-reactivity and false positivity, notably between Babesia and Plasmodium spp., cannot be definitively excluded and have implications for testing in other settings.


Subject(s)
Babesia microti/growth & development , Babesiosis/epidemiology , Malaria, Falciparum/epidemiology , Plasmodium falciparum/growth & development , Antibodies, Protozoan/chemistry , Babesia microti/immunology , Babesiosis/blood , Babesiosis/diagnosis , Babesiosis/parasitology , Child, Preschool , Coinfection , Cross Reactions , Dried Blood Spot Testing , Enzyme-Linked Immunosorbent Assay , Erythrocytes/parasitology , Female , Humans , Infant , Infant, Newborn , Malaria, Falciparum/blood , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Male , Pilot Projects , Plasmodium falciparum/immunology , Tanzania/epidemiology
18.
Immunity ; 47(6): 1197-1209.e10, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29195810

ABSTRACT

Antibodies against the NANP repeat of circumsporozoite protein (CSP), the major surface antigen of Plasmodium falciparum (Pf) sporozoites, can protect from malaria in animal models but protective humoral immunity is difficult to induce in humans. Here we cloned and characterized rare affinity-matured human NANP-reactive memory B cell antibodies elicited by natural Pf exposure that potently inhibited parasite transmission and development in vivo. We unveiled the molecular details of antibody binding to two distinct protective epitopes within the NANP repeat. NANP repeat recognition was largely mediated by germline encoded and immunoglobulin (Ig) heavy-chain complementarity determining region 3 (HCDR3) residues, whereas affinity maturation contributed predominantly to stabilizing the antigen-binding site conformation. Combined, our findings illustrate the power of exploring human anti-CSP antibody responses to develop tools for malaria control in the mammalian and the mosquito vector and provide a molecular basis for the structure-based design of next-generation CSP malaria vaccines.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Immunity, Humoral , Immunoglobulin Heavy Chains/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/immunology , Female , Gene Expression , Humans , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/chemistry , Immunologic Memory , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Mice , Models, Molecular , Plasmodium berghei/immunology , Plasmodium falciparum/immunology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sporozoites/chemistry , Sporozoites/immunology
19.
Proc Natl Acad Sci U S A ; 114(48): E10438-E10445, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29138320

ABSTRACT

Acquired resistance against antimalarial drugs has further increased the need for an effective malaria vaccine. The current leading candidate, RTS,S, is a recombinant circumsporozoite protein (CSP)-based vaccine against Plasmodium falciparum that contains 19 NANP repeats followed by a thrombospondin repeat domain. Although RTS,S has undergone extensive clinical testing and has progressed through phase III clinical trials, continued efforts are underway to enhance its efficacy and duration of protection. Here, we determined that two monoclonal antibodies (mAbs 311 and 317), isolated from a recent controlled human malaria infection trial exploring a delayed fractional dose, inhibit parasite development in vivo by at least 97%. Crystal structures of antibody fragments (Fabs) 311 and 317 with an (NPNA)3 peptide illustrate their different binding modes. Notwithstanding, one and three of the three NPNA repeats adopt similar well-defined type I ß-turns with Fab311 and Fab317, respectively. Furthermore, to explore antibody binding in the context of P. falciparum CSP, we used negative-stain electron microscopy on a recombinant shortened CSP (rsCSP) construct saturated with Fabs. Both complexes display a compact rsCSP with multiple Fabs bound, with the rsCSP-Fab311 complex forming a highly organized helical structure. Together, these structural insights may aid in the design of a next-generation malaria vaccine.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/therapy , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Antigens, Protozoan/isolation & purification , Antigens, Protozoan/therapeutic use , Clinical Trials, Phase II as Topic , Crystallography, X-Ray , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Humans , Malaria Vaccines/chemistry , Malaria Vaccines/therapeutic use , Malaria, Falciparum/immunology , Mice , Mice, Inbred C57BL , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/isolation & purification , Protozoan Proteins/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Repetitive Sequences, Amino Acid/immunology , Structure-Activity Relationship
20.
Nat Commun ; 8(1): 1568, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29146922

ABSTRACT

The Plasmodium falciparum Pfs25 protein (Pfs25) is a leading malaria transmission-blocking vaccine antigen. Pfs25 vaccination is intended to elicit antibodies that inhibit parasite development when ingested by Anopheles mosquitoes during blood meals. The Pfs25 three-dimensional structure has remained elusive, hampering a molecular understanding of its function and limiting immunogen design. We report six crystal structures of Pfs25 in complex with antibodies elicited by immunization via Pfs25 virus-like particles in human immunoglobulin loci transgenic mice. Our structural findings reveal the fine specificities associated with two distinct immunogenic sites on Pfs25. Importantly, one of these sites broadly overlaps with the epitope of the well-known 4B7 mouse antibody, which can be targeted simultaneously by antibodies that target a non-overlapping site to additively increase parasite inhibition. Our molecular characterization of inhibitory antibodies informs on the natural disposition of Pfs25 on the surface of ookinetes and provides the structural blueprints to design next-generation immunogens.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Blocking/chemistry , Antibodies, Blocking/immunology , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/immunology , Crystallography, X-Ray , Female , HEK293 Cells , Humans , Immunization , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Male , Mice, Transgenic , Plasmodium falciparum/metabolism , Protein Binding , Protein Domains , Protozoan Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...