Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Agric Food Chem ; 70(41): 13062-13070, 2022 Oct 19.
Article in English | MEDLINE | ID: mdl-35834180

ABSTRACT

Gut microbiota modulation might constitute a mechanism mediating the effects of beer on health. In this randomized, double-blinded, two-arm parallel trial, 22 healthy men were recruited to drink 330 mL of nonalcoholic beer (0.0% v/v) or alcoholic beer (5.2% v/v) daily during a 4-week follow-up period. Blood and faecal samples were collected before and after the intervention period. Gut microbiota was analyzed by 16S rRNA gene sequencing. Drinking nonalcoholic or alcoholic beer daily for 4 weeks did not increase body weight and body fat mass and did not changed significantly serum cardiometabolic biomarkers. Nonalcoholic and alcoholic beer increased gut microbiota diversity which has been associated with positive health outcomes and tended to increase faecal alkaline phosphatase activity, a marker of intestinal barrier function. These results suggest the effects of beer on gut microbiota modulation are independent of alcohol and may be mediated by beer polyphenols.


Subject(s)
Beer , Gastrointestinal Microbiome , Male , Humans , Beer/analysis , RNA, Ribosomal, 16S/genetics , Alkaline Phosphatase , Biomarkers
2.
Clin Infect Dis ; 69(9): 1509-1516, 2019 10 15.
Article in English | MEDLINE | ID: mdl-30629148

ABSTRACT

BACKGROUND: Malaria in pregnancy has major impacts on mother and child health. To complement existing interventions, such as intermittent preventive treatment and use of impregnated bed nets, we developed a malaria vaccine candidate with the aim of reducing sequestration of asexual "blood-stage" parasites in the placenta, the major virulence mechanism. METHODS: The vaccine candidate PAMVAC is based on a recombinant fragment of VAR2CSA, the Plasmodium falciparum protein responsible for binding to the placenta via chondroitin sulfate A (CSA). Healthy, adult malaria-naive volunteers were immunized with 3 intramuscular injections of 20 µg (n = 9) or 50 µg (n = 27) PAMVAC, adjuvanted with Alhydrogel or glucopyranosyl lipid adjuvant in stable emulsion (GLA-SE) or in a liposomal formulation with QS21 (GLA-LSQ). Allocation was random and double blind. The vaccine was given every 4 weeks. Volunteers were observed for 6 months following last immunization. RESULTS: All PAMVAC formulations were safe and well tolerated. A total of 262 adverse events (AEs) occurred, 94 (10 grade 2 and 2 grade 3) at least possibly related to the vaccine. No serious AEs occurred. Distribution and severity of AEs were similar in all arms. PAMVAC was immunogenic in all participants. PAMVAC-specific antibody levels were highest with PAMVAC-GLA-SE. The antibodies inhibited binding of VAR2CSA expressing P. falciparum-infected erythrocytes to CSA in a standardized functional assay. CONCLUSIONS: PAMVAC formulated with Alhydrogel or GLA-based adjuvants was safe, well tolerated, and induced functionally active antibodies. Next, PAMVAC will be assessed in women before first pregnancies in an endemic area. CLINICAL TRIALS REGISTRATION: EudraCT 2015-001827-21; ClinicalTrials.gov NCT02647489.


Subject(s)
Malaria Vaccines/therapeutic use , Adult , Aluminum Hydroxide/chemistry , Chondroitin Sulfates/metabolism , Double-Blind Method , Female , Humans , Injections, Intramuscular , Liposomes/chemistry , Malaria Vaccines/administration & dosage , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Pregnancy , Young Adult
3.
J Infect Dis ; 216(12): 1601-1610, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29029247

ABSTRACT

Malaria during pregnancy constitutes a large health problem in areas of endemicity. The World Health Organization recommends that interventions are initiated at the first antenatal visit, and these improve pregnancy outcomes. This study evaluated fetal growth by ultrasonography and birth outcomes in women who were infected prior to the first antenatal visit (gestational age, <120 days) and not later in pregnancy. Compared with uninfected controls, women with early Plasmodium falciparum exposure had retarded intrauterine growth between gestational ages of 212 and 253 days (difference between means, 107 g [95% confidence interval {CI}, 26-188]; P = .0099) and a shorter pregnancy duration (difference between means, 6.6 days [95% CI, 1.0-112.5]; P = .0087). The birth weight (difference between means, 221 g [95% CI, 6-436]; P = .044) and the placental weight (difference between means, 84 g [95% CI, 18-150]; P = .013) at term were also reduced. The study suggests that early exposure to P. falciparum, which is not targeted for prevention by current control strategies, has a profound impact on fetal growth, pregnancy duration, and placental weight at term.


Subject(s)
Fetal Development , Malaria, Falciparum/complications , Pregnancy Complications, Infectious/pathology , Pregnancy Outcome , Adolescent , Adult , Female , Humans , Longitudinal Studies , Placenta/pathology , Pregnancy , Ultrasonography , Young Adult
4.
Vaccine ; 35(8): 1140-1147, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28131394

ABSTRACT

The malaria parasite Plasmodium falciparum presents antigens on the infected erythrocyte surface that bind human receptors expressed on the vascular endothelium. The VAR2CSA mediated binding to a distinct chondroitin sulphate A (CSA) is a crucial step in the pathophysiology of placental malaria and the CSA binding region of VAR2CSA has been identified as a promising vaccine target against placental malaria. Here we designed adenovirus encoded virus-like particles (VLP) by co-encoding Simian Immunodeficiency Virus (SIV) gag and VAR2CSA. The VAR2CSA antigen was fused to the transmembrane (TM) and cytoplasmic tail (CT) domains of either the envelope protein of mouse mammary tumour virus (MMTV) or the hemagglutinin (HA) of influenza A. For a non-VLP incorporation control, a third design was made where VAR2CSA was expressed without TM-CT domains. In the primary immunogenicity study in Balb/c mice, VAR2CSA fused to HA TM-CT was significantly superior in inducing ID1-ID2a specific antibodies after the first immunization. A sequential study was performed to include a comparison to the soluble VAR2CSA protein vaccine, which has entered a phase I clinical trial (NCT02647489). The results revealed the induction of higher antibody responses and increased inhibition of parasite binding to CSA using either VAR2CSA HA TM-CT or VAR2CSA MMTV TM-CT as priming vaccines for protein double-boost immunizations, compared to protein prime-double boost regimen. Analysis of pooled serum samples on peptide arrays revealed a unique targeting of several epitopes in mice that had been primed with VAR2CSA HA TM-CT. Consequently, modification of VLP anchors is an important point of optimization in virus-encoded retroviral VLP-based vaccines, and adenovirus VLPs boosted by recombinant proteins offer hope of increasing the levels of protective VAR2CSA specific antibodies.


Subject(s)
Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Vaccines, Virus-Like Particle/administration & dosage , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Female , Gene Products, gag/chemistry , Gene Products, gag/genetics , Gene Products, gag/immunology , Genetic Vectors/chemistry , Genetic Vectors/immunology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunization , Malaria Vaccines/biosynthesis , Malaria Vaccines/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mammary Tumor Virus, Mouse , Mice , Mice, Inbred BALB C , Placenta/chemistry , Placenta/immunology , Placenta/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , Pregnancy , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Simian Immunodeficiency Virus , Vaccines, Virus-Like Particle/biosynthesis , Vaccines, Virus-Like Particle/genetics
5.
Vaccine ; 35(4): 610-618, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28012775

ABSTRACT

BACKGROUND: Malaria in pregnancy is associated with significant morbidity in pregnant women and their offspring. Plasmodium falciparum infected erythrocytes (IE) express VAR2CSA that mediates binding to chondroitin sulphate A (CSA) in the placenta. Two VAR2CSA-based vaccines for placental malaria are in clinical development. The purpose of this study was to evaluate the robustness and comparability of binding inhibition assays used in the clinical development of placental malaria vaccines. METHODS: The ability of sera from animals immunised with different VAR2CSA constructs to inhibit IE binding to CSA was investigated in three in vitro assays using 96-well plates, petri dishes, capillary flow and an ex vivo placental perfusion assay. RESULTS: The inter-assay variation was not uniform between assays and ranged from above ten-fold in the flow assay to two-fold in the perfusion assay. The intra-assay variation was highest in the petri dish assay. A positive correlation between IE binding avidity and the level of binding after antibody inhibition in the petri dish assay indicate that high avidity IE binding is more difficult to inhibit. The highest binding inhibition sensitivity was found in the 96-well and petri dish assays compared to the flow and perfusion assays where binding inhibition required higher antibody titers. CONCLUSIONS: The inhibitory capacity of antibodies is not easily translated between assays and the high sensitivity of the 96-well and petri dish assays stresses the need for comparing serial dilutions of serum. Furthermore, IE binding avidity must be in the same range when comparing data from different days. There was an overall concordance in the capacity of antibody-mediated inhibition, when comparing the in vitro assays with the perfusion assay, which more closely represents in vivo conditions. Importantly the ID1-ID2a protein in a liposomal formulation, currently in a phase I trial, effectively induced antibodies that inhibited IE adhesion in placental tissue.


Subject(s)
Antibodies, Protozoan/blood , Cell Adhesion , Chondroitin Sulfates/metabolism , Cytological Techniques/methods , Erythrocytes/physiology , Malaria, Falciparum/prevention & control , Placenta Diseases/prevention & control , Animals , Antigens, Protozoan/immunology , Drug Discovery/methods , Erythrocytes/parasitology , Female , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Mice, Inbred C57BL , Pregnancy , Rabbits , Rats, Wistar , Reproducibility of Results
6.
PLoS Pathog ; 12(8): e1005831, 2016 08.
Article in English | MEDLINE | ID: mdl-27556547

ABSTRACT

During placental malaria, Plasmodium falciparum infected erythrocytes sequester in the placenta, causing health problems for both the mother and fetus. The specific adherence is mediated by the VAR2CSA protein, which binds to placental chondroitin sulfate (CS) on chondroitin sulfate proteoglycans (CSPGs) in the placental syncytium. However, the identity of the CSPG core protein and the cellular impact of the interaction have remain elusive. In this study we identified the specific CSPG core protein to which the CS is attached, and characterized its exact placental location. VAR2CSA pull-down experiments using placental extracts from whole placenta or syncytiotrophoblast microvillous cell membranes showed three distinct CSPGs available for VAR2CSA adherence. Further examination of these three CSPGs by immunofluorescence and proximity ligation assays showed that syndecan-1 is the main receptor for VAR2CSA mediated placental adherence. We further show that the commonly used placental choriocarcinoma cell line, BeWo, express a different set of proteoglycans than those present on placental syncytiotrophoblast and may not be the most biologically relevant model to study placental malaria. Syncytial fusion of the BeWo cells, triggered by forskolin treatment, caused an increased expression of placental CS-modified syndecan-1. In line with this, we show that rVAR2 binding to placental CS impairs syndecan-1-related Src signaling in forskolin treated BeWo cells, but not in untreated cells.


Subject(s)
Antigens, Protozoan/metabolism , Chondroitin Sulfates/metabolism , Malaria, Falciparum/parasitology , Placenta/parasitology , Pregnancy Complications, Infectious/parasitology , Syndecan-1/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Malaria, Falciparum/metabolism , Mass Spectrometry , Microscopy, Confocal , Microscopy, Electron, Transmission , Placenta/metabolism , Plasmodium falciparum , Pregnancy , Pregnancy Complications, Infectious/metabolism
7.
Sens Biosensing Res ; 9: 23-30, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27441183

ABSTRACT

In clinical oncology, diagnosis and evaluation of optimal treatment strategies are mostly based on histopathological examination combined with immunohistochemical (IHC) expression analysis of cancer-associated antigens in formalin fixed paraffin-embedded (FFPE) tissue biopsies. However, informative IHC analysis depends on both the specificity and affinity of the binding reagent, which are inherently difficult to quantify in situ. Here we describe a label-free method that allows for the direct and real-time assessment of molecular binding kinetics in situ on FFPE tissue specimens using quartz crystal microbalance (QCM) enabled biosensor technology. We analysed the interaction between the rVAR2 protein and its placental-like chondroitin sulfate (pl-CS) receptor in primary human placenta tissue and in breast and prostate tumour specimens in situ. rVAR2 interacted with FFPE human placenta and cancer tissue with an affinity in the nanomolar range, and showed no detectable interaction with pl-CS negative normal tissue. We further validated the method by including analysis with the androgen receptor N-20 antibody (anti-AR). As the KD value produced by this method is independent of the number of epitopes available, this readout offers a quantitative and unbiased readout for in situ binding-avidity and amount of binding epitopes. In summary, this method adds a new and important dimension to classical IHC-based molecular pathology by adding information about the binding characteristics in biologically relevant conditions. This can potentially be used to select optimal biologics for diagnostic and for therapeutic applications as well as guide the development of novel high affinity binding drugs.

8.
Malar J ; 15(1): 292, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27230523

ABSTRACT

BACKGROUND: Placental malaria occurs when Plasmodium falciparum infected erythrocytes sequester in the placenta. Placental parasite isolates bind to chondroitin sulphate A (CSA) by expression of VAR2CSA on the surface of infected erythrocytes, but may sequester by other VAR2CSA mediated mechanisms, such as binding to immunoglobulins. Furthermore, other parasite antigens have been associated with placental malaria. These findings have important implications for placental malaria vaccine design. The objective of this study was to adapt and describe a biologically relevant model of parasite adhesion in intact placental tissue. RESULTS: The ex vivo placental perfusion model was modified to study adhesion of infected erythrocytes binding to CSA, endothelial protein C receptor (EPCR) or a transgenic parasite where P. falciparum erythrocyte membrane protein 1 expression had been shut down. Infected erythrocytes expressing VAR2CSA accumulated in perfused placental tissue whereas the EPCR binding and the transgenic parasite did not. Soluble CSA and antibodies specific against VAR2CSA inhibited binding of infected erythrocytes. CONCLUSION: The ex vivo model provides a novel way of studying receptor-ligand interactions and antibody mediated inhibition of binding in placental malaria.


Subject(s)
Cell Adhesion , Erythrocytes/physiology , Erythrocytes/parasitology , Malaria, Falciparum/pathology , Placenta Diseases/pathology , Placenta/pathology , Placenta/parasitology , Female , Humans , Malaria, Falciparum/parasitology , Models, Theoretical , Placenta Diseases/parasitology , Plasmodium falciparum , Pregnancy
9.
J Nanobiotechnology ; 14: 30, 2016 Apr 27.
Article in English | MEDLINE | ID: mdl-27117585

ABSTRACT

BACKGROUND: Virus-like particles (VLPs) represent a significant advance in the development of subunit vaccines, combining high safety and efficacy. Their particulate nature and dense repetitive subunit organization makes them ideal scaffolds for display of vaccine antigens. Traditional approaches for VLP-based antigen display require labor-intensive trial-and-error optimization, and often fail to generate dense antigen display. Here we utilize the split-intein (SpyTag/SpyCatcher) conjugation system to generate stable isopeptide bound antigen-VLP complexes by simply mixing of the antigen and VLP components. RESULTS: Genetic fusion of SpyTag or SpyCatcher to the N-terminus and/or C-terminus of the Acinetobacter phage AP205 capsid protein resulted in formation of stable, nonaggregated VLPs expressing one SpyCatcher, one SpyTag or two SpyTags per capsid protein. Mixing of spy-VLPs with eleven different vaccine antigens fused to SpyCatcher or SpyTag resulted in formation of antigen-VLP complexes with coupling efficiencies (% occupancy of total VLP binding sites) ranging from 22-88 %. In mice, spy-VLP vaccines presenting the malaria proteins Pfs25 or VAR2CSA markedly increased antibody titer, affinity, longevity and functional efficacy compared to corresponding vaccines employing monomeric proteins. The spy-VLP vaccines also effectively broke B cell self-tolerance and induced potent and durable antibody responses upon vaccination with cancer or allergy-associated self-antigens (PD-L1, CTLA-4 and IL-5). CONCLUSIONS: The spy-VLP system constitutes a versatile and rapid method to develop highly immunogenic VLP-based vaccines. Our data provide proof-of-concept for the technology's ability to present complex vaccine antigens to the immune system and elicit robust functional antibody responses as well as to efficiently break B cell self-tolerance. The spy-VLP-system may serve as a generic tool for the cost-effective development of effective VLP-vaccines against both infectious- and non-communicable diseases and could facilitate rapid and unbiased screening of vaccine candidate antigens.


Subject(s)
Vaccines, Virus-Like Particle/immunology , Acinetobacter/immunology , Animals , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , Bacteriophages/immunology , Capsid Proteins/immunology , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Vaccination/methods
10.
PLoS One ; 10(11): e0143071, 2015.
Article in English | MEDLINE | ID: mdl-26599509

ABSTRACT

Placental malaria caused by Plasmodium falciparum is a major cause of mortality and severe morbidity. Clinical testing of a soluble protein-based vaccine containing the parasite ligand, VAR2CSA, has been initiated. VAR2CSA binds to the human receptor chondroitin sulphate A (CSA) and is responsible for sequestration of Plasmodium falciparum infected erythrocytes in the placenta. It is imperative that a vaccine against malaria in pregnancy, if administered to women before they become pregnant, can induce a strong and long lasting immune response. While most soluble protein-based vaccines have failed during clinical testing, virus-like particle (VLP) based vaccines (e.g., the licensed human papillomavirus vaccines) have demonstrated high efficacy, suggesting that the spatial assembly of the vaccine antigen is a critical parameter for inducing an optimal long-lasting protective immune response. We have developed a VLP vaccine display platform by identifying regions of the HPV16 L1 coat protein where a biotin acceptor site (AviTagTM) can be inserted without compromising VLP-assembly. Subsequent biotinylation of Avi-L1 VLPs allow us to anchor monovalent streptavidin (mSA)-fused proteins to the biotin, thereby obtaining a dense and repetitive VLP-display of the vaccine antigen. The mSA-VAR2CSA antigen was delivered on the Avi-L1 VLP platform and tested in C57BL/6 mice in comparison to two soluble protein-based vaccines consisting of naked VAR2CSA and mSA-VAR2CSA. The mSA-VAR2CSA Avi-L1 VLP and soluble mSA-VAR2CSA vaccines induced higher antibody titers than the soluble naked VAR2CSA vaccine after three immunizations. The VAR2CSA Avi-L1 VLP vaccine induced statistically significantly higher endpoint titres compared to the soluble mSA-VAR2CSA vaccine, after 1st and 2nd immunization; however, this difference was not statistically significant after 3rd immunization. Importantly, the VLP-VAR2CSA induced antibodies were functional in inhibiting the binding of parasites to CSA. This study demonstrates that the described Avi-L1 VLP-platform may serve as a versatile system for facilitating optimal VLP-display of large and complex vaccine antigens.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Placenta/parasitology , Virion/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Biotinylation , Blotting, Western , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Malaria, Falciparum/blood , Malaria, Falciparum/immunology , Mice, Inbred C57BL , Molecular Sequence Data , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/metabolism , Parasites/immunology , Pregnancy , Reproducibility of Results , Ultracentrifugation , Vaccination
11.
PLoS One ; 10(9): e0135406, 2015.
Article in English | MEDLINE | ID: mdl-26327283

ABSTRACT

The disease caused by Plasmodium falciparum (Pf) involves different clinical manifestations that, cumulatively, kill hundreds of thousands every year. Placental malaria (PM) is one such manifestation in which Pf infected erythrocytes (IE) bind to chondroitin sulphate A (CSA) through expression of VAR2CSA, a parasite-derived antigen. Protection against PM is mediated by antibodies that inhibit binding of IE in the placental intervillous space. VAR2CSA is a large antigen incompatible with large scale recombinant protein expression. Vaccines based on sub-units encompassing the functionally constrained receptor-binding domains may, theoretically, circumvent polymorphisms, reduce the risk of escape-mutants and induce cross-reactive antibodies. However, the sub-unit composition and small differences in the borders, may lead to exposure of novel immuno-dominant antibody epitopes that lead to non-functional antibodies, and furthermore influence the folding, stability and yield of expression. Candidate antigens from the pre-clinical development expressed in High-Five insect cells using the baculovirus expression vector system were transitioned into the Drosophila Schneider-2 cell (S2) expression-system compliant with clinical development. The functional capacity of antibodies against antigens expressed in High-Five cells or in S2 cells was equivalent. This enabled an extensive down-selection of S2 insect cell-expressed antigens primarily encompassing the minimal CSA-binding region of VAR2CSA. In general, we found differential potency of inhibitory antibodies against antigens with the same borders but of different var2csa sequences. Likewise, we found that subtle size differences in antigens of the same sequence gave varying levels of inhibitory antibodies. The study shows that induction of a functional response against recombinant subunits of the VAR2CSA antigen is unpredictable, demonstrating the need for large-scale screening in order to identify antigens that induce a broadly strain-transcending antibody response.


Subject(s)
Antibody Formation/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Placenta/parasitology , Animals , Cross Reactions/immunology , Drosophila melanogaster/metabolism , Female , Humans , Malaria, Falciparum/immunology , Placenta/immunology , Plasmodium falciparum/immunology , Pregnancy , Protein Engineering/methods , Recombinant Proteins
12.
Infect Immun ; 81(2): 487-95, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23208604

ABSTRACT

Pregnancy malaria is caused by Plasmodium falciparum-infected erythrocytes that adhere to the placental receptor chondroitin sulfate A (CSA) and sequester in the placenta; women become resistant to pregnancy malaria as they acquire antiadhesion antibodies that target surface proteins of placental parasites. VAR2CSA, a member of the P. falciparum EMP1 variant surface antigen family, is the leading candidate for a pregnancy malaria vaccine. Because VAR2CSA is a high-molecular-weight protein, a vaccine based on the full-length protein may not be feasible. An alternative approach has been to develop a vaccine targeting individual Duffy binding-like (DBL) domains. In this study, a consortium of laboratories under the Pregnancy Malaria Initiative compared the functional activity of antiadhesion antibodies elicited by different VAR2CSA domains and variants produced in prokaryotic and eukaryotic expression systems. Antisera were initially tested against laboratory lines of maternal parasites, and the most promising reagents were evaluated in the field against fresh placental parasite samples. Recombinant proteins expressed in Escherichia coli elicited antibody levels similar to those expressed in eukaryotic systems, as did the two allelic forms of the DBL4 and DBL5 domains. The procedures developed for this head-to-head comparison will be useful for future evaluation and down-selection of malaria vaccine immunogens.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Placenta/immunology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Animals , Chondroitin Sulfates/immunology , Cohort Studies , Female , Humans , Immune Sera/immunology , Immunoglobulin G/immunology , Longitudinal Studies , Malaria Vaccines/pharmacology , Malaria, Falciparum/prevention & control , Pregnancy , Pregnancy Complications, Parasitic/prevention & control , Rats , Recombinant Proteins/immunology
13.
PLoS One ; 7(9): e43663, 2012.
Article in English | MEDLINE | ID: mdl-22970138

ABSTRACT

Malaria during pregnancy in Plasmodium falciparum endemic regions is a major cause of mortality and severe morbidity. VAR2CSA is the parasite ligand responsible for sequestration of Plasmodium falciparum infected erythrocytes to the receptor chondroitin sulfate A (CSA) in the placenta and is the leading candidate for a placental malaria vaccine. Antibodies induced in rats against the recombinant DBL4ε domain of VAR2CSA inhibit the binding of a number of laboratory and field parasite isolates to CSA. In this study, we used a DBL4ε peptide-array to identify epitopes targeted by DBL4ε-specific antibodies that inhibit CSA-binding of infected erythrocytes. We identified three regions of overlapping peptides which were highly antigenic. One peptide region distinguished itself particularly by showing a clear difference in the binding profile of highly parasite blocking IgG compared to the IgG with low capacity to inhibit parasite adhesion to CSA. This region was further characterized and together these results suggest that even though antibodies against the synthetic peptides which cover this region did not recognize native protein, the results using the mutant domain suggest that this linear epitope might be involved in the induction of inhibitory antibodies induced by the recombinant DBL4ε domain.


Subject(s)
Antigens, Protozoan/chemistry , Epitopes, B-Lymphocyte/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/immunology , Antibody Formation/immunology , Antibody Specificity/immunology , Antigens, Protozoan/immunology , Cell Adhesion , Epitope Mapping , Epitopes, B-Lymphocyte/chemistry , Erythrocytes/parasitology , Female , Humans , Immune Sera/immunology , Linear Models , Models, Molecular , Molecular Sequence Data , Multivariate Analysis , Mutant Proteins/chemistry , Mutant Proteins/immunology , Parasites/immunology , Peptides/chemistry , Peptides/immunology , Plasmodium falciparum/cytology , Plasmodium falciparum/immunology , Pregnancy , Protein Structure, Tertiary , Rats , Sequence Alignment
14.
J Biol Chem ; 287(28): 23332-45, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22570492

ABSTRACT

Malaria is a major global health problem. Pregnant women are susceptible to infection regardless of previously acquired immunity. Placental malaria is caused by parasites capable of sequestering in the placenta. This is mediated by VAR2CSA, a parasite antigen that interacts with chondroitin sulfate A (CSA). One vaccine strategy is to block this interaction with VAR2CSA-specific antibodies. It is a priority to define a small VAR2CSA fragment that can be used in an adhesion blocking vaccine. In this, the obvious approach is to define regions of VAR2CSA involved in receptor binding. It has been shown that full-length recombinant VAR2CSA binds specifically to CSA with nanomolar affinity, and that the CSA-binding site lies in the N-terminal part of the protein. In this study we define the minimal binding region by truncating VAR2CSA and analyzing CSA binding using biosensor technology. We show that the core CSA-binding site lies within the DBL2X domain and parts of the flanking interdomain regions. This is in contrast to the idea that single domains do not possess the structural requirements for specific CSA binding. Small-angle x-ray scattering measurements enabled modeling of VAR2CSA and showed that the CSA-binding DBL2X domain is situated in the center of the structure. Mutating classic sulfate-binding sites in VAR2CSA, along with testing dependence of ionic interactions, suggest that the CSA binding is not solely dependent on the sulfated CSA structure. Based on these novel PfEMP1 structure-function studies, we have constructed a small VAR2CSA antigen that has the capacity to induce highly adhesion-blocking antibodies.


Subject(s)
Antigens, Protozoan/immunology , Chondroitin Sulfates/immunology , Malaria, Falciparum/immunology , Placenta/immunology , Plasmodium falciparum/immunology , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Binding Sites/genetics , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Female , Host-Parasite Interactions , Humans , Immune Sera/immunology , Immune Sera/metabolism , Immunization , Kinetics , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Models, Molecular , Mutation , Placenta/metabolism , Placenta/parasitology , Plasmodium falciparum/physiology , Pregnancy , Pregnancy Complications, Parasitic , Protein Binding , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Scattering, Small Angle , X-Ray Diffraction
15.
PLoS One ; 6(3): e17942, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21464946

ABSTRACT

BACKGROUND: In Plasmodium falciparum malaria endemic areas placental malaria (PM) is an important complication of malaria. The recurrence of malaria in primigravidae women irrespective of acquired protection during childhood is caused by the interaction between the parasite-expressed VAR2CSA antigen and chondroitin sulfate A (CSA) in the placental intervillous space and lack of protective antibodies. PM impairs fetal development mainly by excessive inflammation processes. After infections during pregnancy women acquire immunity to PM conferred by antibodies against VAR2CSA. Ideally, a vaccine against PM will induce antibody-mediated immune responses that block the adhesion of infected erythrocytes (IE) in the placenta. PRINCIPAL FINDINGS: We have previously shown that antibodies raised in rat against individual domains of VAR2CSA can block IE binding to CSA. In this study we have immunized mice, rats and rabbits with each individual domain and the full-length protein corresponding to the FCR3 VAR2CSA variant. We found there is an inherently higher immunogenicity of C-terminal domains compared to N-terminally located domains. This was irrespective of whether antibodies were induced against single domains or the full-length protein. Species-specific antibody responses were also found, these were mainly directed against single domains and not the full-length VAR2CSA protein. CONCLUSIONS/SIGNIFICANCE: Binding inhibitory antibodies appeared to be against conformational B-cell epitopes. Non-binding inhibitory antibodies reacted highly against the C-terminal end of the VAR2CSA molecule especially the highly polymorphic DBL6ε domain. Differential species-specific induction of antibody responses may allow for more direct analysis of functional versus non-functional B-cell epitopes.


Subject(s)
Antigens, Protozoan/immunology , Immunoglobulin G/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Placenta/immunology , Placenta/parasitology , Plasmodium falciparum/immunology , Animals , Antibodies, Protozoan/immunology , Antibody Formation/immunology , Antigens, Protozoan/chemistry , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Erythrocytes/parasitology , Female , Humans , Malaria, Falciparum/parasitology , Mice , Pregnancy , Protein Array Analysis , Protein Structure, Tertiary , Rabbits , Rats , Recombinant Proteins/immunology , Species Specificity
16.
J Biol Chem ; 286(18): 15908-17, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21398524

ABSTRACT

Malaria during pregnancy is a major health problem for African women. The disease is caused by Plasmodium falciparum malaria parasites, which accumulate in the placenta by adhering to chondroitin sulfate A (CSA). The interaction between infected erythrocytes and the placental receptor is mediated by a parasite expressed protein named VAR2CSA. A vaccine protecting pregnant women against placental malaria should induce antibodies inhibiting the interaction between VAR2CSA and CSA. Much effort has been put into defining the part of the 350 kDa VAR2CSA protein that is responsible for binding. It has been shown that full-length recombinant VAR2CSA binds specifically to CSA with high affinity, however to date no sub-fragment of VAR2CSA has been shown to interact with CSA with similar affinity or specificity. In this study, we used a biosensor technology to examine the binding properties of a panel of truncated VAR2CSA proteins. The experiments indicate that the core of the CSA-binding site is situated in three domains, DBL2X-CIDR(PAM) and a flanking domain, located in the N-terminal part of VAR2CSA. Furthermore, recombinant VAR2CSA subfragments containing this region elicit antibodies with high parasite adhesion blocking activity in animal immunization experiments.


Subject(s)
Chondroitin Sulfates/chemistry , Peptide Mapping , Plasmodium falciparum/chemistry , Animals , Antigens, Protozoan , Biosensing Techniques/methods , Chondroitin Sulfates/genetics , Chondroitin Sulfates/immunology , Chondroitin Sulfates/metabolism , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/parasitology , Female , Humans , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria Vaccines/metabolism , Malaria, Falciparum/epidemiology , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Malaria, Falciparum/prevention & control , Placenta/immunology , Placenta/metabolism , Placenta/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Plasmodium falciparum/metabolism , Pregnancy , Pregnancy Complications, Parasitic/epidemiology , Pregnancy Complications, Parasitic/genetics , Pregnancy Complications, Parasitic/immunology , Pregnancy Complications, Parasitic/metabolism , Pregnancy Complications, Parasitic/prevention & control , Protein Binding , Protein Structure, Tertiary , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
17.
Vaccine ; 29(3): 437-43, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-21075162

ABSTRACT

Malaria during pregnancy is a major cause of intra-uterine growth-retardation and infant death in sub-Saharan Africa. Ideally, this could be prevented by a vaccine delivered before the first pregnancy. Antibodies against domain DBL4ɛ from VAR2CSA has been shown to inhibit adhesion of laboratory isolates to the placental receptor chondroitin sulfate A. In this study, the binding inhibitory efficacy of IgG elicited by two different DBL4ɛ recombinant proteins was tested on a panel of fresh clinical isolates from pregnant women living in Benin and Tanzania. The most promising recombinant protein elicited antibodies with similar efficacy as pooled plasma from immune multi-gravid African women.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Cell Adhesion , DNA Repair Enzymes/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/immunology , Pregnancy Complications, Infectious/immunology , Transcription Factors/metabolism , Benin , Female , Humans , Immunoglobulin G/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Pregnancy , Recombinant Proteins/immunology , Tanzania
18.
J Mol Biol ; 397(3): 826-34, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20109466

ABSTRACT

Plasmodium falciparum malaria remains one of the world's leading causes of human suffering and poverty. Each year, the disease takes 1-3 million lives, mainly in sub-Saharan Africa. The adhesion of infected erythrocytes (IEs) to vascular endothelium or placenta is the key event in the pathogenesis of severe P. falciparum infection. In pregnant women, the parasites express a single and unique member of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) family named VAR2CSA, which is associated with the ability of the IEs to adhere specifically to chondroitin sulphate A (CSA) in the placenta. Several Duffy-binding-like domains from VAR2CSA molecules have been shown in vitro to bind to CSA, but it has also been demonstrated that Duffy-binding-like domains from PfEMP1 proteins other than VAR2CSA can bind CSA. In addition, the specificity of the binding of VAR2CSA domains to glycosaminoglycans does not match that of VAR2CSA-expressing IEs. This has led to speculation that the domains of native VAR2CSA need to come together to form a specific binding site or that VAR2CSA might bind to CSA through a bridging molecule. Here, we describe the expression and purification of the complete extracellular region of VAR2CSA secreted at high yields from insect cells. Using surface plasmon resonance, we demonstrate that VAR2CSA alone binds with nanomolar affinity to human chondroitin sulphate proteoglycan and with significantly weaker affinity to other glycosaminoglycans, showing a specificity similar to that observed for IEs. Antibodies raised against full-length VAR2CSA completely inhibit recombinant VAR2CSA binding, as well as parasite binding to chondroitin sulphate proteoglycan. This is the first study to describe the successful production and functionality of a full-length PfEMP1. The specificity of the binding and anti-adhesion potency of induced IgG, together with high-yield production, encourages the use of full-length PfEMP1 in vaccine development strategies.


Subject(s)
Antibodies, Protozoan/immunology , Antibodies, Protozoan/metabolism , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Chondroitin Sulfates/metabolism , Erythrocytes/metabolism , Animals , Antigens, Protozoan/genetics , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Erythrocytes/parasitology , Flow Cytometry , Glycosaminoglycans/metabolism , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Surface Plasmon Resonance
19.
Malar J ; 9: 11, 2010 Jan 11.
Article in English | MEDLINE | ID: mdl-20064234

ABSTRACT

BACKGROUND: Malaria caused by Plasmodium falciparum can result in several different syndromes with severe clinical consequences for the about 200 million individuals infected each year. During pregnancy, women living in endemic areas become susceptible to malaria due to lack of antibodies against a unique P. falciparum membrane protein, named VAR2CSA. This antigen is not expressed in childhood infections, since it binds chondroitin sulphate A (CSA) expressed on the intervillous space in the placenta. A vaccine appears possible because women acquire protective antibodies hindering sequestration in the placenta as a function of parity. A challenge for vaccine development is to design small constructs of this large antigen, which can induce broadly protective antibodies. It has previously been shown that one domain of VAR2CSA, DBL4-FCR3, induces parasite adhesion-blocking antibodies. In this study, it is demonstrated that other domains of VAR2CSA also can induce antibodies with inhibitory activity. METHODS: All VAR2CSA domains from the 3D7 and HB3 parasites were produced in Baculovirus-transfected insect cells. Groups of three rats per protein were immunized and anti-sera were tested for surface reactivity against infected erythrocytes expressing FCR3 VAR2CSA and for the ability to inhibit FCR3CSA parasite adhesion to CSA. The fine specificity of the immune sera was analysed by VAR2CSA peptide arrays. RESULTS: Inhibitory antibodies were induced by immunization with DBL3-HB3 T1 and DBL1-3D7. However, unlike the previously characterised DBL4-FCR3 response the inhibitory response against DBL1-3D7 and DBL3-HB3 T1 was poorly reproduced in the second rounds of immunizations. CONCLUSION: It is possible to induce parasite adhesion-blocking antibodies when immunizing with a number of different VAR2CSA domains. This indicates that the CSA binding site in VAR2CSA is comprised of epitopes from different domains.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Cell Adhesion/immunology , Malaria Vaccines/immunology , Animals , Antigens, Protozoan/genetics , Baculoviridae/genetics , Female , Genetic Vectors , Humans , Insecta , Malaria Vaccines/genetics , Pregnancy , Protein Structure, Tertiary , Rats , Rats, Wistar , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
20.
Infect Immun ; 77(6): 2482-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19307213

ABSTRACT

In areas of endemicity pregnancy-associated malaria is an important cause of maternal anemia, stillbirth, and delivery of low-birth-weight children. The syndrome is precipitated by the accumulation of Plasmodium falciparum-infected erythrocytes in the placenta, mediated through an interaction between a parasite protein expressed on erythrocytes named variant surface antigen 2-chondroitin sulfate A (VAR2CSA) and CSA on syncytiotrophoblasts. VAR2CSA is a large polymorphic protein consisting of six Duffy binding-like (DBL), domains and with current constraints on recombinant protein production it is not possible to produce entire VAR2CSA recombinant proteins. Furthermore, the presence of polymorphisms has raised the question of whether it is feasible to define VAR2CSA antigens eliciting broadly protective antibodies. Thus, the challenge for vaccine development is to define smaller parts of the molecule which induce antibodies that inhibit CSA binding of different parasite strains. In this study, we produced a large panel of VAR2CSA proteins and raised antibodies against these antigens. We show that antibodies against the DBL4 domain effectively inhibit parasite binding. As the inhibition was not limited to homologous parasite strains, it seems feasible to base a protective malaria vaccine on a single VAR2CSA DBL domain.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Cell Adhesion/immunology , Plasmodium falciparum/immunology , Trophoblasts/parasitology , Animals , Antigens, Protozoan/genetics , Humans , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Plasmodium falciparum/physiology , Rats , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...