Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
NPJ Vaccines ; 9(1): 10, 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38184681

ABSTRACT

The receptor-binding domain, region II, of the Plasmodium vivax Duffy binding protein (PvDBPII) binds the Duffy antigen on the reticulocyte surface to mediate invasion. A heterologous vaccine challenge trial recently showed that a delayed dosing regimen with recombinant PvDBPII SalI variant formulated with adjuvant Matrix-MTM reduced the in vivo parasite multiplication rate (PMR) in immunized volunteers challenged with the Thai P. vivax isolate PvW1. Here, we describe extensive analysis of the polyfunctional antibody responses elicited by PvDBPII immunization and identify immune correlates for PMR reduction. A classification algorithm identified antibody features that significantly contribute to PMR reduction. These included antibody titre, receptor-binding inhibitory titre, dissociation constant of the PvDBPII-antibody interaction, complement C1q and Fc gamma receptor binding and specific IgG subclasses. These data suggest that multiple immune mechanisms elicited by PvDBPII immunization are likely to be associated with protection and the immune correlates identified could guide the development of an effective vaccine for P. vivax malaria. Importantly, all the polyfunctional antibody features that correlated with protection cross-reacted with both PvDBPII SalI and PvW1 variants, suggesting that immunization with PvDBPII should protect against diverse P. vivax isolates.

2.
Sci Rep ; 13(1): 13904, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37626150

ABSTRACT

The invasion of reticulocytes by Plasmodium vivax merozoites is dependent on the interaction of the Plasmodium vivax Duffy Binding Protein (PvDBP) with the Duffy antigen receptor for chemokines (DARC). The N-terminal cysteine-rich region II of PvDBP (PvDBPII), which binds DARC, is a leading P. vivax malaria vaccine candidate. Here, we have evaluated the immunogenicity of recombinant PvDBPII formulated with the adjuvants Matrix-M and GLA-SE in mice. Analysis of the antibody responses revealed comparable ELISA recognition titres as well as similar recognition of native PvDBP in P. vivax schizonts by immunofluorescence assay. Moreover, antibodies elicited by the two adjuvant formulations had similar functional properties such as avidity, isotype profile and inhibition of PvDBPII-DARC binding. Furthermore, the anti-PvDBPII antibodies were able to block the interaction of DARC with the homologous PvDBPII SalI allele as well as the heterologous PvDBPII PvW1 allele from a Thai clinical isolate that is used for controlled human malaria infections (CHMI). The cross-reactivity of these antibodies with PvW1 suggest that immunization with the PvDBPII SalI strain should neutralize reticulocyte invasion by the challenge P. vivax strain PvW1.


Subject(s)
Malaria, Vivax , Vaccines , Humans , Animals , Mice , Plasmodium vivax , Carrier Proteins , Adjuvants, Immunologic , Adjuvants, Pharmaceutic , Antibodies , Malaria, Vivax/prevention & control
3.
Sci Transl Med ; 15(704): eadf1782, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37437014

ABSTRACT

There are no licensed vaccines against Plasmodium vivax. We conducted two phase 1/2a clinical trials to assess two vaccines targeting P. vivax Duffy-binding protein region II (PvDBPII). Recombinant viral vaccines using chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) vectors as well as a protein and adjuvant formulation (PvDBPII/Matrix-M) were tested in both a standard and a delayed dosing regimen. Volunteers underwent controlled human malaria infection (CHMI) after their last vaccination, alongside unvaccinated controls. Efficacy was assessed by comparisons of parasite multiplication rates in the blood. PvDBPII/Matrix-M, given in a delayed dosing regimen, elicited the highest antibody responses and reduced the mean parasite multiplication rate after CHMI by 51% (n = 6) compared with unvaccinated controls (n = 13), whereas no other vaccine or regimen affected parasite growth. Both viral-vectored and protein vaccines were well tolerated and elicited expected, short-lived adverse events. Together, these results support further clinical evaluation of the PvDBPII/Matrix-M P. vivax vaccine.


Subject(s)
Malaria , Parasites , Humans , Animals , Plasmodium vivax , Vaccination
4.
Front Immunol ; 14: 1193079, 2023.
Article in English | MEDLINE | ID: mdl-38299155

ABSTRACT

We have previously reported primary endpoints of a clinical trial testing two vaccine platforms for the delivery of Plasmodium vivax malaria DBPRII: viral vectors (ChAd63, MVA), and protein/adjuvant (PvDBPII with 50µg Matrix-M™ adjuvant). Delayed boosting was necessitated due to trial halts during the pandemic and provides an opportunity to investigate the impact of dosing regimens. Here, using flow cytometry - including agnostic definition of B cell populations with the clustering tool CITRUS - we report enhanced induction of DBPRII-specific plasma cell and memory B cell responses in protein/adjuvant versus viral vector vaccinees. Within protein/adjuvant groups, delayed boosting further improved B cell immunogenicity compared to a monthly boosting regimen. Consistent with this, delayed boosting also drove more durable anti-DBPRII serum IgG. In an independent vaccine clinical trial with the P. falciparum malaria RH5.1 protein/adjuvant (50µg Matrix-M™) vaccine candidate, we similarly observed enhanced circulating B cell responses in vaccinees receiving a delayed final booster. Notably, a higher frequency of vaccine-specific (putatively long-lived) plasma cells was detected in the bone marrow of these delayed boosting vaccinees by ELISPOT and correlated strongly with serum IgG. Finally, following controlled human malaria infection with P. vivax parasites in the DBPRII trial, in vivo growth inhibition was observed to correlate with DBPRII-specific B cell and serum IgG responses. In contrast, the CD4+ and CD8+ T cell responses were impacted by vaccine platform but not dosing regimen and did not correlate with in vivo growth inhibition in a challenge model. Taken together, our DBPRII and RH5 data suggest an opportunity for protein/adjuvant dosing regimen optimisation in the context of rational vaccine development against pathogens where protection is antibody-mediated.


Subject(s)
Malaria, Vivax , Vaccines , Humans , Plasmodium falciparum , Bone Marrow , Antigens, Protozoan , Adjuvants, Immunologic , Malaria, Vivax/prevention & control , Immunoglobulin G
5.
medRxiv ; 2022 May 30.
Article in English | MEDLINE | ID: mdl-35664997

ABSTRACT

Background: There are no licensed vaccines against Plasmodium vivax , the most common cause of malaria outside of Africa. Methods: We conducted two Phase I/IIa clinical trials to assess the safety, immunogenicity and efficacy of two vaccines targeting region II of P. vivax Duffy-binding protein (PvDBPII). Recombinant viral vaccines (using ChAd63 and MVA vectors) were administered at 0, 2 months or in a delayed dosing regimen (0, 17, 19 months), whilst a protein/adjuvant formulation (PvDBPII/Matrix-M™) was administered monthly (0, 1, 2 months) or in a delayed dosing regimen (0, 1, 14 months). Delayed regimens were due to trial halts during the COVID-19 pandemic. Volunteers underwent heterologous controlled human malaria infection (CHMI) with blood-stage P. vivax parasites at 2-4 weeks following their last vaccination, alongside unvaccinated controls. Efficacy was assessed by comparison of parasite multiplication rate (PMR) in blood post-CHMI, modelled from parasitemia measured by quantitative polymerase-chain-reaction (qPCR). Results: Thirty-two volunteers were enrolled and vaccinated (n=16 for each vaccine). No safety concerns were identified. PvDBPII/Matrix-M™, given in the delayed dosing regimen, elicited the highest antibody responses and reduced the mean PMR following CHMI by 51% (range 36-66%; n=6) compared to unvaccinated controls (n=13). No other vaccine or regimen impacted parasite growth. In vivo growth inhibition of blood-stage P. vivax correlated with functional antibody readouts of vaccine immunogenicity. Conclusions: Vaccination of malaria-naïve adults with a delayed booster regimen of PvDBPII/ Matrix-M™ significantly reduces the growth of blood-stage P. vivax . Funded by the European Commission and Wellcome Trust; VAC069, VAC071 and VAC079 ClinicalTrials.gov numbers NCT03797989 , NCT04009096 and NCT04201431 .

6.
Vaccines (Basel) ; 9(7)2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34358198

ABSTRACT

A fusion chimeric vaccine comprising multiple protective domains of different blood-stage Plasmodium falciparum antigens is perhaps necessary for widening the protective immune responses and reducing the morbidity caused by the disease. Here we continue to build upon the prior work of developing a recombinant fusion chimera protein, His-tagged PfMSP-Fu24, by producing it as a tag-free recombinant protein. In this study, tag-free recombinant PfMSPFu24 (rFu24) was expressed in Escherichia coli, and the soluble protein was purified using a three-step purification involving ammonium sulphate precipitation followed by 2-step ion exchange chromatography procedures and shown that it was highly immunogenic with the human-compatible adjuvant Alhydrogel. We further investigated two dipeptides, phenylalanine-α, ß-dehydrophenylalanine (FΔF) and Leucine-α, ß-dehydrophenylalanine (LΔF) based hydrogels as effective delivery platforms for rFu24. These dipeptides self-assembled spontaneously to form a highly stable hydrogel under physiological conditions. rFu24 was efficiently entrapped in both the F∆F and L∆F hydrogels, and the three-dimensional (3D) mesh-like structures of the hydrogels remained intact after the entrapment of the antigen. The two hydrogels significantly stimulated rFu24-specific antibody titers, and the sera from the immunized mice showed an invasion inhibitory activity comparable to that of Alhydrogel. Easily synthesized dipeptide hydrogels can be used as an effective antigen delivery platform to induce immune responses.

7.
NPJ Vaccines ; 3: 48, 2018.
Article in English | MEDLINE | ID: mdl-30302285

ABSTRACT

Reticulocyte invasion by Plasmodium vivax requires interaction of the Duffy-binding protein (PvDBP) with host Duffy antigen receptor for chemokines (DARCs). The binding domain of PvDBP maps to a cysteine-rich region referred to as region II (PvDBPII). Blocking this interaction offers a potential path to prevent P. vivax blood-stage growth and P. vivax malaria. This forms the rationale for development of a vaccine based on PvDBPII. Here we report results of a Phase I randomized trial to evaluate the safety and immunogenicity of recombinant PvDBPII formulated with glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE). Thirty-six malaria-naive, healthy Indian male subjects aged 18-45 years were assigned into three cohorts corresponding to doses of 10, 25 and 50 µg of PvDBPII formulated with 5 µg of GLA-SE. Each cohort included nine PvDBPII/GLA-SE vaccinees and three hepatitis B control vaccine recipients. Each subject received the assigned vaccine intramuscularly on days 0, 28 and 56, and was followed up till day 180. No serious AE was reported and PvDBPII/GLA-SE was well-tolerated and safe. Analysis by ELISA showed that all three doses of PvDBPII elicited antigen-specific binding-inhibitory antibodies. The 50 µg dose elicited antibodies against PvDBPII that had the highest binding-inhibitory titres and were most persistent. Importantly, the antibody responses were strain transcending and blocked receptor binding of diverse PvDBP alleles. These results support further clinical development of PvDBPII/GLA-SE to evaluate efficacy against sporozoite or blood-stage challenge in controlled human malaria infection (CHMI) models and against natural P. vivax challenge in malaria endemic areas.

8.
Malar J ; 15: 457, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27604988

ABSTRACT

BACKGROUND: Immunological characterization of potential blood-stage malaria antigens would be a valuable strategy in the development of an effective vaccine. Identifying B and CD4(+) T cell epitopes will be important in understanding the nature of immune response. A previous study has shown that Plasmodium falciparum apical asparagine-rich protein (PfAARP) stimulates immune response and induces potent invasion-inhibitory antibodies. Antibodies to PfAARP provide synergistic effects in inhibition of parasite invasion when used in combination with antibodies to other antigens. In the present study, an attempt was made to identify B cell and CD4(+) T cell epitopes of PfAARP. METHODS: Balb/c mice were immunized with recombinant PfAARP and both cellular and humoral responses were analysed at various time points. Computerized databases [immune epitope database (IEDB) and B cell epitope prediction (BCEPred)] were used to predict epitope sequences within PfAARP and predicted peptides were synthesized. In addition, nine 18 amino acid, long-overlapping peptides spanning the entire length of PfAARP were synthesized. Using these peptides, B cell and CD4(+) T cell responses in PfAARP immunized mice were measured by ELISA and ELISPOT assays. RESULTS: Here, it is demonstrated that immunization of mice with PfAARP induced long-lasting, high-titre antibodies (4 months post immunization). Also, the recombinant protein was effective in inducing a pronounced Th1 type of immune response quantified by IFN-γ ELISA and ELISPOT. It was found that the predicted peptides did not represent the immunogenic regions of PfAARP. However, of the nine overlapping peptides, three peptides (peptides 3, 5 and 7) were strongly recognized by PfAARP-immunized sera and represented B cell epitopes. Also, peptide 3 elicited IFN- γ response, suggesting it to be a T-cell epitope. CONCLUSIONS: Induction of long-lasting humoral and cellular response on PfAARP immunization in mice underscores its possible use as a blood-stage malaria vaccine candidate. Mapping of immunogenic regions may help in designing fusion chimera containing immunologically relevant regions of other vaccine target antigens and/or for multi-component vaccine candidates.


Subject(s)
Antigens, Protozoan/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Epitope Mapping , Female , Immunity, Cellular , Immunity, Humoral , Mice, Inbred BALB C , Time Factors
10.
PLoS One ; 10(4): e0117820, 2015.
Article in English | MEDLINE | ID: mdl-25927360

ABSTRACT

BACKGROUND: A phase I randomised, controlled, single blind, dose escalation trial was conducted to evaluate safety and immunogenicity of JAIVAC-1, a recombinant blood stage vaccine candidate against Plasmodium falciparum malaria, composed of a physical mixture of two recombinant proteins, PfMSP-1(19), the 19 kD conserved, C-terminal region of PfMSP-1 and PfF2 the receptor-binding F2 domain of EBA175. METHOD: Healthy malaria naïve Indian male subjects aged 18-45 years were recruited from the volunteer database of study site. Fifteen subjects in each cohort, randomised in a ratio of 2:1 and meeting the protocol specific eligibility criteria, were vaccinated either with three doses (10 µg, 25 µg and 50 µg of each antigen) of JAIVAC-1 formulated with adjuvant Montanide ISA 720 or with standard dosage of Hepatitis B vaccine. Each subject received the assigned vaccine in the deltoid muscle of the upper arms on Day 0, Day 28 and Day 180. RESULTS: JAIVAC-1 was well tolerated and no serious adverse event was observed. All JAIVAC-1 subjects sero-converted for PfF2 but elicited poor immune response to PfMSP-1(19). Dose-response relationship was observed between vaccine dose of PfF2 and antibody response. The antibodies against PfF2 were predominantly of IgG1 and IgG3 isotype. Sera from JAIVAC-1 subjects reacted with late schizonts in a punctate pattern in immunofluorescence assays. Purified IgG from JAIVAC-1 sera displayed significant growth inhibitory activity against Plasmodium falciparum CAMP strain. CONCLUSION: Antigen PfF2 should be retained as a component of a recombinant malaria vaccine but PfMSP-1(19) construct needs to be optimised to improve its immunogenicity. TRIAL REGISTRATION: Clinical Trial Registry, India CTRI/2010/091/000301.


Subject(s)
Antigens, Protozoan/administration & dosage , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Merozoite Surface Protein 1/administration & dosage , Plasmodium falciparum/immunology , Protozoan Proteins/administration & dosage , Adolescent , Adult , Antibodies, Protozoan/immunology , Antigens, Protozoan/adverse effects , Antigens, Protozoan/immunology , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Female , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/adverse effects , Hepatitis B Vaccines/immunology , Humans , Immunoglobulin G/immunology , India , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Male , Mannitol/administration & dosage , Mannitol/adverse effects , Mannitol/analogs & derivatives , Merozoite Surface Protein 1/adverse effects , Merozoite Surface Protein 1/immunology , Middle Aged , Oleic Acids/administration & dosage , Oleic Acids/adverse effects , Protozoan Proteins/adverse effects , Protozoan Proteins/immunology
11.
Clin Vaccine Immunol ; 21(6): 886-97, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24789797

ABSTRACT

A Plasmodium falciparum chimeric protein, PfMSP-Fu24, was constructed by genetically coupling immunodominant, conserved regions of two merozoite surface proteins, the 19-kDa region C-terminal region of merozoite surface protein 1 (PfMSP-119) and an 11-kDa conserved region of merozoite surface protein 3 (PfMSP-311), to augment the immunogenicity potential of these blood-stage malaria vaccine candidates. Here we describe an improved, efficient, and scalable process to produce high-quality PfMSP-Fu24. The chimeric protein was produced in Escherichia coli SHuffle T7 Express lysY cells that express disulfide isomerase DsbC. A two-step purification process comprising metal affinity followed by cation exchange chromatography was developed, and we were able to obtain PfMSP-Fu24 with purity above 99% and with a considerable yield of 23 mg/liter. Immunogenicity of PfMSP-Fu24 formulated with several adjuvants, including Adjuplex, Alhydrogel, Adjuphos, Alhydrogel plus glucopyranosyl lipid adjuvant, aqueous (GLA-AF), Adjuphos+GLA-AF, glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE), and Freund's adjuvant, was evaluated. PfMSP-Fu24 formulated with GLA-SE and Freund's adjuvant in mice and with Alhydrogel and Freund's adjuvant in rabbits produced high titers of PfMSP-119 and PfMSP-311-specific functional antibodies. Some of the adjuvant formulations induced inhibitory antibody responses and inhibited in vitro growth of P. falciparum parasites in the presence as well as in the absence of human monocytes. These results suggest that PfMSP-Fu24 can form a constituent of a multistage malaria vaccine.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Recombinant Fusion Proteins/immunology , Adjuvants, Immunologic , Animals , Antibodies, Protozoan/immunology , Antibody Formation/immunology , Antigens, Protozoan/genetics , Drug Evaluation, Preclinical , Escherichia coli/genetics , Female , Humans , Malaria Vaccines/genetics , Malaria, Falciparum/prevention & control , Merozoite Surface Protein 1/genetics , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Rabbits , Recombinant Fusion Proteins/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
12.
Mol Biochem Parasitol ; 191(2): 58-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24080030

ABSTRACT

Subtilisin-like proteases of malaria parasite Plasmodium falciparum (PfSUB1, 2 and 3) are expressed at late asexual blood stages. PfSUB1 and 2 are considered important drug targets due to their essentiality for parasite blood stages and role in merozoite egress and invasion of erythrocytes. We have earlier shown the in vitro serine protease activity of PfSUB3 and its localization at asexual blood stages. In this study, we attempted to identify the biological substrate(s) of PfSUB3 and found parasite profilin (PfPRF) as a substrate of the protease. Eukaryotic profilins are multifunctional proteins with primary role in regulation of actin filament assembly. PfPRF possesses biochemical features of eukaryotic profilins and its rodent ortholog is essential in blood stages. Profilin from related apicomplexan parasite Toxoplasma gondii (TgPRF) is known to be involved in parasite motility, host cell invasion, active egress from host cell, immune evasion and virulence in mice. In this study, mature PfSUB3 proteolysed recombinant PfPRF in a dose-dependent manner in in vitro assays. Recombinant PfPRF was assessed for its proinflammatory activity and found to induce high level of TNF-α and low but significant level of IL-12 from mouse bone marrow-derived dendritic cells. Proteolysis of PfPRF by PfSUB3 is suggestive of the probable role of the protease in the processes of motility, virulence and immune evasion.


Subject(s)
Malaria, Falciparum/parasitology , Plasmodium falciparum/enzymology , Profilins/metabolism , Protozoan Proteins/metabolism , Subtilisin/metabolism , Amino Acid Sequence , Animals , Cell Line , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Humans , Interleukin-12/genetics , Interleukin-12/metabolism , Malaria, Falciparum/genetics , Malaria, Falciparum/metabolism , Mice , Molecular Sequence Data , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Profilins/genetics , Protein Binding , Proteolysis , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Homology, Amino Acid , Subtilisin/chemistry , Subtilisin/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Infect Immun ; 80(2): 633-42, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22104109

ABSTRACT

The 19-kDa carboxyl-terminal fragment of the merozoite surface protein-1 (MSP-1(19)) has been shown to regulate antibody (Ab)-mediated protective immunity to blood-stage malaria infection. But the serological memory to this antigen tends to be short-lived, and little is known of the mechanisms that regulate the formation of B cell memory to MSP-1(19) antigen. We studied the formation of B cell memory response after immunization with the recombinant 19-kDa Plasmodium falciparum merozoite surface protein 1 (PfMSP-1(19)). Immunization with PfMSP-1(19) resulted in delayed increase in germinal center (GC) B cell numbers. This poor GC reaction correlated with short-lived PfMSP-1(19)-specific antibodies in serum and the short life of PfMSP-1(19)-specific plasma cells and memory B cells (MBCs) in spleen and bone marrow. PfMSP-1(19)-specific MBCs were capable of producing antigen (Ag)-specific Ab-secreting cell (ASC) responses that were short-lived following challenge immunization of the immune mice with antigen or transgenic Plasmodium berghei parasite expressing PfMSP-1(19) in place of native P. berghei MSP-1(19) at 8 weeks after the last immunization or following adoptive transfer into naive hosts. However, no protection was achieved in PfMSP-1(19) immune mice or recipient mice with PfMSP-1(19)-specific MBCs following challenge with transgenic P. berghei. Our findings suggest that PfMSP-1(19)-specific IgG production by short-lived plasma cells combined with the poor ability of the PfMSP-1(19)-induced MBCs to maintain the anamnestic IgG responses failed to contribute to protection against infection.


Subject(s)
B-Lymphocytes/physiology , Immunity, Humoral/physiology , Immunologic Memory/physiology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Adoptive Transfer , Animals , Female , Gene Expression Regulation , Immunization , Immunoglobulin G/blood , Immunoglobulin G/classification , Kinetics , Mice , Mice, Inbred BALB C , Plasmodium falciparum/metabolism
14.
Infect Immun ; 78(2): 872-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19933832

ABSTRACT

A chimeric gene, MSP-Fu(24), was constructed by genetically coupling immunodominant, conserved regions of the two leading malaria vaccine candidates, Plasmodium falciparum merozoite surface protein 1 (C-terminal 19-kDa region [PfMSP-1(19)]) and merozoite surface protein 3 (11-kDa conserved region [PfMSP-3(11)]). The recombinant MSP-Fu(24) protein was produced in Escherichia coli cells and purified to homogeneity by a two-step purification process with a yield of approximately 30 mg/liter. Analyses of conformational properties of MSP-Fu(24) using PfMSP-1(19)-specific monoclonal antibody showed that the conformational epitopes of PfMSP-1(19) that may be critical for the generation of the antiparasitic immune response remained intact in the fusion protein. Recombinant MSP-Fu(24) was highly immunogenic in mice and in rabbits when formulated with two different human-compatible adjuvants and induced an immune response against both PfMSP-1(19) and PfMSP-3(11). Purified anti-MSP-Fu(24) antibodies showed invasion inhibition of P. falciparum 3D7 and FCR parasites, and this effect was found to be dependent on antibodies specific for the PfMSP-1(19) component. The protective potential of MSP-Fu(24) was demonstrated by in vitro parasite growth inhibition using an antibody-dependent cell inhibition (ADCI) assay with anti-MSP-Fu(24) antibodies. Overall, the antiparasitic activity was mediated by a combination of growth-inhibitory antibodies generated by both the PfMSP-1(19) and PfMSP-3(11) components of the MSP-Fu(24) protein. The antiparasitic activities elicited by anti-MSP-Fu(24) antibodies were comparable to those elicited by antibodies generated with immunization with a physical mixture of two component antigens, PfMSP-1(19) and PfMSP-3(11). The fusion protein induces a protective immune response with human-compatible adjuvants and may form a part of a multicomponent malaria vaccine.


Subject(s)
Adjuvants, Immunologic , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Merozoite Surface Protein 1/genetics , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Mice , Plasmodium falciparum , Rabbits , Recombinant Proteins/immunology
15.
Mol Cell Proteomics ; 8(9): 2102-18, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19494339

ABSTRACT

The highly co-evolved relationship of parasites and their hosts appears to include modulation of host immune signals, although the molecular mechanisms involved in the host-parasite interplay remain poorly understood. Characterization of these key genes and their cognate proteins related to the host-parasite interplay should lead to a better understanding of this intriguing biological phenomenon. The malaria agent Plasmodium falciparum is predicted to export a cohort of several hundred proteins to remodel the host erythrocyte. However, proteins actively exported by the asexual intracellular parasite beyond the host red blood cell membrane (before merozoite egress) have been poorly investigated so far. Here we used two complementary methodologies, two-dimensional gel electrophoresis/MS and LC-MS/MS, to examine the extracellular secreted antigens at asexual blood stages of P. falciparum. We identified 27 novel antigens exported by P. falciparum in the culture medium of which some showed clustering with highly polymorphic genes on chromosomes, suggesting that they may encode putative antigenic determinants of the parasite. Immunolocalization of four novel secreted proteins confirmed their export beyond the infected red blood cell membrane. Of these, preliminary functional characterization of two novel (Sel1 repeat-containing) parasite proteins, PfSEL1 and PfSEL2 revealed that they down-regulate expression of cell surface Notch signaling molecules in host cells. Also a novel protein kinase (PfEK) and a novel protein phosphatase (PfEP) were found to, respectively, phosphorylate/dephosphorylate parasite-specific proteins in the extracellular culture supernatant. Our study thus sheds new light on malaria parasite extracellular secreted antigens of which some may be essential for parasite development and could constitute promising new drug targets.


Subject(s)
Antigens, Protozoan/analysis , Extracellular Space/metabolism , Life Cycle Stages/immunology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Proteome/analysis , Protozoan Proteins/analysis , Signal Transduction/immunology , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Blotting, Western , Chromatography, Liquid , Computational Biology , Electrophoresis, Gel, Two-Dimensional , Erythrocytes/parasitology , Erythrocytes/ultrastructure , Extracellular Space/immunology , Gene Expression Regulation , Genes, Protozoan , Humans , Life Cycle Stages/genetics , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Mass Spectrometry , Parasites/genetics , Parasites/growth & development , Parasites/immunology , Parasites/ultrastructure , Plasmodium falciparum/genetics , Plasmodium falciparum/ultrastructure , Proteome/immunology , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Subcellular Fractions/metabolism , Transcription, Genetic
16.
J Leukoc Biol ; 84(1): 244-54, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18413701

ABSTRACT

Free plasmodium merozoites released from the parasitized hepatocytes and erythrocytes represent a transitory, extracellular stage in its mammalian host. In this study, we compared the effect of Plasmodium falciparum-free merozoites with infected RBCs (iRBCs) on the maturation of human monocyte-derived dendritic cells (DCs) in vitro. Phagocytosed-free merozoites prevented soluble CD40 ligand (sCD40L)-induced, phenotypic maturation of DCs and secretion of IL-12p70 but enhanced IL-10 production and primed, naive CD4+ cells to produce a high level of IL-10 compared with IFN-gamma. Free merozoites augmented sCD40L-induced ERK1/2 activation, and inhibition of ERK1/2 with its inhibitor PD98059 markedly abrogated IL-10 production and rescued IL-12 production. Therefore, the molecular mechanisms by which free merozoites antagonized sCD40L-induced DC maturation appeared to involve the activation of the ERK pathway. In contrast, phagocytosed iRBCs by itself induced DCs to semi-maturation, responded to CD40 signaling by maturing and secreting increased levels of TNF-alpha, IL-6, and also IL-12p70, and led to a pronounced, proinflammatory response by the allogenic CD4+ T cells. iRBCs regulate CD40-induced p38MAPK. Studies using inhibitors selective for p38MAPK (SB203580) showed that p38MAPK played an essential role in the maturation and function of DCs. Our results reveal the ability of free merozoites and iRBCs to distinctly alter the sCD40L-induced DC functioning by regulating the activation of the MAPK pathway that can inactivate or exacerbate immune responses to promote their survival and the development of parasite-specific pathologies.


Subject(s)
CD40 Ligand/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Erythrocytes/parasitology , Merozoites/immunology , Monocytes/cytology , Plasmodium falciparum/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cell Polarity/drug effects , Cytokines/biosynthesis , Erythrocytes/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Malaria, Falciparum/immunology , Merozoites/drug effects , Monocytes/drug effects , Phagocytosis/drug effects , Plasmodium falciparum/drug effects , Solubility/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Vaccine ; 25(28): 5166-74, 2007 Jul 09.
Article in English | MEDLINE | ID: mdl-17544179

ABSTRACT

An effective malaria vaccine will probably require the delivery of multiple antigens that induce several layers of immunity. Malaria antigens expressed on the surface and in apical organelles of blood-stage merozoites are potential vaccine candidates given their importance in the invasion of erythrocytes. The present study examined the kinetics of humoral response in BALB/c mice following immunization with combination of two blood-stage Plasmodium vivax invasion related molecules, the N-terminal, cysteine-rich region II of P. vivax Duffy binding protein (PvRII) and the 19kDa C-terminal region of merozoite surface protein 1 (PvMSP1(19)) formulated with Montanide ISA 720 and alhydrogel. Immunization with combination of recombinant PvRII and PvMSP1(19) formulated with the Montanide ISA 720 elicited higher antibody titer compared to the alhydrogel formulation. In case of both the adjuvants tested, combination of PvRII and PvMSP1(19) did not result in suppression of antibody response against either antigen when compared to immunization with individual antigens alone. Analysis of IgG subclasses showed that combination of both the recombinant proteins induced a mixed Th1/Th2-type response with almost all IgG subtypes being expressed in equivalent amount. Antibodies elicited against PvRII showed significant inhibitory effect on the binding of PvRII to recombinant Duffy antigen receptor for chemokines (DARC) in an in vitro binding assay. The results of the present study provide a rationale for a combination vaccine against P. vivax malaria based on PvMSP1(19) and PvRII.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Antibody Formation/immunology , Antigens, Protozoan/genetics , Blotting, Western , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Immunization/methods , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Male , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/immunology , Mice , Mice, Inbred BALB C , Plasmodium vivax/genetics , Protozoan Proteins/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Recombinant Proteins/immunology , Time Factors
18.
Vaccine ; 25(5): 806-13, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17045706

ABSTRACT

Malaria parasites require specific receptor-ligand interactions to invade host erythrocytes. The 175 kDa Plasmodium falciparum erythrocyte binding antigen (EBA-175) binds sialic acid residues on glycophorin A to mediate erythrocyte invasion. The amino-terminal, conserved, cysteine-rich region of EBA-175, referred to as F2, contains receptor-binding sites. We propose to develop a recombinant malaria vaccine based on region F2. Recombinant P. falciparum region F2 (PfF2) was expressed in Escherichia coli, purified from inclusion bodies under denaturing conditions by metal affinity chromatography, renatured by oxidative refolding and purified further by ion-exchange and gel filtration chromatography. Recombinant PfF2 was characterized and shown to be pure, homogeneous and functionally active in that it binds human erythrocytes with specificity. The immunogenicity of recombinant PfF2 formulated with three human compatible adjuvants, namely, Montanide ISA720, AS02A and alum was tested in mice. All the formulations tested elicited high titer antibodies that block erythrocyte invasion in vitro. The AS02 formulation yielded sera with the highest end-point ELISA titers followed by Montanide ISA720 and alum. Analysis of cellular immune responses indicated that all formulations resulted in significant splenocyte proliferation. Analysis of cytokines secreted by proliferating splenocytes indicated that all the adjuvant formulations tested induced Th1 type responses. These results suggest that recombinant PfF2 formulated with human compatible adjuvants is immunogenic and can elicit high titer invasion inhibitory antibodies providing support for further clinical development of this promising vaccine candidate.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Protozoan Proteins/immunology , Vaccines, Synthetic/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Protozoan/chemistry , Binding Sites , Cytokines/biosynthesis , Malaria Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Protein Folding , Protozoan Proteins/chemistry , Vaccination , Vaccines, Synthetic/administration & dosage
19.
Infect Immun ; 72(10): 5775-82, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15385477

ABSTRACT

The 42- and 19-kDa C-terminal fragments of merozoite surface protein 1 (MSP-1(42) and MSP-1(19), respectively) are both promising blood-stage vaccine candidate antigens. At present, it is not clear which of the two antigens will be more suitable for inclusion in a cocktail malaria vaccine. In the present study, we expressed the two C-terminal fragments of Plasmodium vivax MSP-1 (PvMSP-1) in an Escherichia coli expression system and purified them by using a rapid two-step protocol. Both of the products were recognized by monoclonal antibodies against PvMSP-1 as well as by immune sera from several individuals exposed to P. vivax. We analyzed and compared the immunological responses to recombinant PvMSP-1(19) and PvMSP-1(42) in mice by using six different adjuvant formulations. Moderate to high antibody responses were observed with both of the antigens in different adjuvant formulations. Surprisingly, alum, which is generally considered to be a poor adjuvant for recombinant malaria antigens, was found to be as good an adjuvant as Montanide ISA 720, ASO2A, and other adjuvant formulations. Most adjuvant formulations induced high levels of immunoglobulin G1 (IgG1), followed by IgG3 and IgG2. Lymphocytes from animals in the PvMSP-1(42)- and PvMSP-1(19)-immunized groups showed proliferative responses upon stimulation with the respective antigens, and high levels of interleukin-4 (IL-4), IL-5, and gamma interferon were detected in the culture supernatants. Immunodepletion studies with sera from mice immunized with these two antigens showed that while immunization with PvMSP-1(42) does produce a PvMSP-1(19)-specific response, a substantial portion is also focused on structures in PvMSP-1(42) not represented by the epidermal growth factor-like domains of PvMSP-1(19). These findings may have implications for the design of MSP-1-based vaccine constructs.


Subject(s)
Escherichia coli/genetics , Merozoite Surface Protein 1/chemistry , Merozoite Surface Protein 1/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Plasmodium vivax/immunology , Adjuvants, Immunologic , Animals , Antibodies, Protozoan/analysis , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Humans , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Immunoglobulin Isotypes/analysis , Immunoglobulin Isotypes/immunology , Malaria Vaccines/immunology , Malaria, Vivax/immunology , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/isolation & purification , Mice , Mice, Inbred BALB C , Molecular Weight , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
20.
Vaccine ; 22(27-28): 3727-37, 2004 Sep 09.
Article in English | MEDLINE | ID: mdl-15315853

ABSTRACT

Plasmodium vivax Duffy binding protein (PvDBP) binds the Duffy blood group antigen as the obligate receptor for erythrocyte invasion. We have tested in mice the immunogenicity of recombinant P. vivax region II (PvRII), the receptor-binding domain of PvDBP, formulated with five adjuvants, namely, Montanide ISA720, AS02A, alum, QS21 and MF59. All the formulations elicited high titer antibodies, with Montanide ISA720 and AS02A yielding the highest titers followed by MF59, QS21 and alum. Sera raised against PvRII formulated with AS02A and Montanide ISA720 followed by alum were most effective at blocking PvRII binding to erythrocytes in a functional assay. Analysis of cellular immune responses indicated that all adjuvant groups induced significant interferon-gamma, with alum being the highest interferon-gamma inducer. These results suggest that recombinant PvRII formulated with human compatible adjuvants is immunogenic in small animal models and that Montanide ISA720, AS02A and alum perform better than MF59 and QS21 in terms of their ability to elicit high titer binding inhibitory antibodies.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Mannitol/analogs & derivatives , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Receptors, Cell Surface/immunology , Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Animals , Antibodies, Protozoan/analysis , Antibodies, Protozoan/biosynthesis , COS Cells , Chemistry, Pharmaceutical , Chlorocebus aethiops , Chromatography, Gel , Chromatography, Ion Exchange , Cytokines/analysis , Cytokines/biosynthesis , Duffy Blood-Group System , Endotoxins/analysis , Endotoxins/biosynthesis , Enzyme-Linked Immunosorbent Assay , Erythrocytes/metabolism , Erythrocytes/parasitology , Escherichia coli/metabolism , Fluorescent Antibody Technique , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Mannitol/pharmacology , Mice , Mice, Inbred BALB C , Oleic Acids/pharmacology , Protein Folding , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...