Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Protein Sci ; 33(1): e4852, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38059674

ABSTRACT

The circumsporozoite protein (CSP) is the main surface antigen of the Plasmodium sporozoite (SPZ) and forms the basis of the currently only licensed anti-malarial vaccine (RTS,S/AS01). CSP uniformly coats the SPZ and plays a pivotal role in its immunobiology, in both the insect and the vertebrate hosts. Although CSP's N-terminal domain (CSPN ) has been reported to play an important role in multiple CSP functions, a thorough biophysical and structural characterization of CSPN is currently lacking. Here, we present an alternative method for the recombinant production and purification of CSPN from Plasmodium falciparum (PfCSPN ), which provides pure, high-quality protein preparations with high yields. Through an interdisciplinary approach combining in-solution experimental methods and in silico analyses, we provide strong evidence that PfCSPN is an intrinsically disordered region displaying some degree of compaction.


Subject(s)
Antimalarials , Malaria Vaccines , Malaria, Falciparum , Humans , Plasmodium falciparum/genetics , Malaria Vaccines/chemistry , Malaria Vaccines/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/chemistry
2.
Trop Biomed ; 40(1): 37-44, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-37356002

ABSTRACT

Circumsporozoite protein (CSP) central repeat region is one of the main target regions of the RTS,S/AS01 vaccine for falciparum infection as it consists of immunodominant B cell epitopes. However, there is a lack of study for P. knowlesi CSP central repeat region. This study aims to characterise the CSP repeat motifs of P. knowlesi isolates in Peninsular Malaysia. CSP repeat motifs of 64 P. knowlesi isolates were identified using Rapid Automatic Detection and Alignment of Repeats (RADAR). Antigenicity of the repeat motifs and linear B cell epitopes were predicted using VaxiJen 2.0, BepiPred-2.0 and BCPred, respectively. A total of 35 dominant repeat motifs were identified. The repeat motif "AGQPQAQGDGANAGQPQAQGDGAN" has the highest repeat frequency (n=15) and antigenicity index of 1.7986. All the repeat regions were predicted as B cell epitopes. In silico approaches revealed that all repeat motifs were antigenic and consisted of B cell epitopes which could be designed as knowlesi malaria vaccine.


Subject(s)
Malaria Vaccines , Plasmodium knowlesi , Epitopes, B-Lymphocyte , Protozoan Proteins , Malaria Vaccines/genetics , Malaria Vaccines/metabolism , Plasmodium falciparum
3.
Biotechnol Bioeng ; 119(10): 2784-2793, 2022 10.
Article in English | MEDLINE | ID: mdl-35822551

ABSTRACT

Virus-like particles (VLPs) induce strong humoral and cellular responses and have formed the basis of some currently licensed vaccines. Here, we present the method used for the production of R21, a VLP-based anti-sporozoite malaria vaccine, under current Clinical Good Manufacturing Practice regulations (cGMP). Previous preclinical studies in BALB/c mice showed that R21 produced almost complete protection against sporozoite challenge with transgenic Plasmodium berghei parasites. Here, we have modified the preclinical production process to enable the production of sufficient quantities of highly pure, clinical-grade material for use in human clinical trials. The R21 construct was re-engineered to include a C-tag to allow affinity-based separation from the major contaminant alcohol oxidase 1 (AOX 1, ~74 kDa). To our knowledge, this is the first use of C-tag technology to purify a VLP vaccine candidate for use in human clinical trials. The R21 vaccine has shown high-level efficacy in an African Phase IIb trial, and multiple clinical trials are underway to assess the safety and efficacy of the vaccine. Our findings support the future use of C-tag platform technologies to enable cGMP-compliant biomanufacturing of high purity yeast-expressed VLP-based vaccines for early phase clinical trials when clinical grade material is required in smaller quantities in a quick time frame.


Subject(s)
Malaria Vaccines , Malaria , Saccharomycetales , Vaccines, Virus-Like Particle , Viral Vaccines , Animals , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/metabolism , Humans , Malaria/prevention & control , Malaria Vaccines/genetics , Malaria Vaccines/metabolism , Mice , Mice, Inbred BALB C , Pichia/genetics
4.
Biochem Biophys Res Commun ; 534: 86-93, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33316544

ABSTRACT

This work describes a methodology for developing a minimal, subunit-based, multi-epitope, multi-stage, chemically-synthesised, anti-Plasmodium falciparum malaria vaccine. Some modified high activity binding peptides (mHABPs) derived from functionally relevant P. falciparum MSP, RH5 and AMA-1 conserved amino acid regions (cHABPs) for parasite binding to and invasion of red blood cells (RBC) were selected. They were highly immunogenic as assessed by indirect immunofluorescence (IFA) and Western blot (WB) assays and protective immune response-inducers against malarial challenge in the Aotus monkey experimental model. NetMHCIIpan 4.0 was used for predicting peptide-Aotus/human major histocompatibility class II (MHCII) binding affinity in silico due to the similarity between Aotus and human immune system molecules; ∼50% of Aotus MHCII allele molecules have a counterpart in the human immune system, being Aotus-specific, whilst others enabled recognition of their human counterparts. Some peptides' 1H-NMR-assessed structural conformation was determined to explain residue modifications in mHABPs inducing secondary structure changes. These directly influenced immunological behaviour, thereby highlighting the relationship with MHCII antigen presentation. The data obtained in such functional, immunological, structural and predictive approach suggested that some of these peptides could be excellent components of a fully-protective antimalarial vaccine.


Subject(s)
Erythrocytes/parasitology , Malaria Vaccines/pharmacology , Plasmodium falciparum/pathogenicity , Animals , Antigens, Protozoan/chemistry , Aotidae , Carrier Proteins/chemistry , Epitopes , Erythrocytes/drug effects , Histocompatibility Antigens Class II/metabolism , Host-Parasite Interactions/drug effects , Humans , Magnetic Resonance Spectroscopy , Malaria Vaccines/immunology , Malaria Vaccines/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Peptides/immunology , Peptides/metabolism , Protozoan Proteins/chemistry , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology
5.
Sci Rep ; 9(1): 5923, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976034

ABSTRACT

Malaria symptoms and pathology are initiated by invasion of host erythrocytes by Plasmodium merozoites in a complex process that involves interactions between parasite and host erythrocyte proteins. Erythrocyte invasion presents attractive targets for malaria vaccine and drug development. Recently it was observed that antibodies against PfMSA180 (PF3D7_1014100) are associated with protection from symptomatic malaria, suggesting that this protein is a target of naturally acquired protective antibodies. Here we characterize PfMSA180, a ~170 kDa merozoite surface antigen that is potentially involved in erythrocyte invasion. PfMSA180 synthesized by the wheat germ cell-free system was used to raise antibodies in rabbits. Growth inhibition assays revealed that parasite invasion is inhibited by antibodies to the PfMSA180 C-terminal region, which contains an erythrocyte-binding domain. Surface plasmon resonance analysis showed that PfMSA180 specifically interacts with human erythrocyte integrin associated protein (CD47), suggesting that PfMSA180 plays a role during merozoite invasion of erythrocytes. Polymorphism analysis revealed that pfmsa180 is highly conserved among field isolates. We show that naturally acquired PfMSA180-specific antibodies responses are associated with protective immunity in a malaria-exposed Thai population. In sum, the data presented here supports further evaluation of the conserved erythrocyte-binding C-terminal region of PfMSA180 as an asexual blood-stage malaria vaccine candidate.


Subject(s)
CD47 Antigen/metabolism , Erythrocytes/metabolism , Malaria Vaccines/metabolism , Malaria, Falciparum/prevention & control , Merozoites/metabolism , Plasmodium falciparum/immunology , Protozoan Proteins/metabolism , Animals , Antibodies, Protozoan/immunology , Antibody Formation , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Merozoites/immunology , Mice , Plasmodium falciparum/metabolism , Rabbits
6.
Cytometry A ; 95(7): 722-725, 2019 07.
Article in English | MEDLINE | ID: mdl-30919583

ABSTRACT

A 26-color staining panel was developed to profile human antigen-specific T cells in an intracellular cytokine staining (ICS) assay using peptide pools to various antigens of interest. In addition to multiple functional markers, the panel includes differentiation/activation markers and markers to assess γδ, mucosal-associated invariant T, and NK T cells as well as conventional NK cells. Panel optimization was performed using previously cryopreserved PBMC from healthy adults, and then, expression of key functional markers in the panel was cross-validated against a validated ICS assay used in the HIV Vaccine Trials Network (HVTN). The panel is currently being used to evaluate the responses to tuberculosis and malaria vaccine candidates in volunteers from different geographic areas. © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Subject(s)
Flow Cytometry/methods , Killer Cells, Natural/immunology , Natural Killer T-Cells/immunology , T-Lymphocytes/immunology , AIDS Vaccines/immunology , AIDS Vaccines/metabolism , Adult , Antigens/metabolism , Cytokines/metabolism , Fluorescent Dyes/chemistry , Humans , Immunologic Memory , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/metabolism , Malaria Vaccines/immunology , Malaria Vaccines/metabolism , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tuberculosis/immunology , Tuberculosis/metabolism
7.
Am J Trop Med Hyg ; 99(4): 827-832, 2018 10.
Article in English | MEDLINE | ID: mdl-30141395

ABSTRACT

In August 2017, the National Institute of Allergy and Infectious Diseases convened a meeting, entitled "Understanding the Liver-Stage Biology of Malaria Parasites to Enable and Accelerate the Development of a Highly Efficacious Vaccine," to discuss the needs and strategies to develop a highly efficacious, whole organism-based vaccine targeting the liver stage of malaria parasites. It was concluded that attenuated sporozoite platforms have proven to be promising approaches, and that late-arresting sporozoites could potentially offer greater vaccine performance than early-arresting sporozoites against malaria. New knowledge and emerging technologies have made the development of late-arresting sporozoites feasible. Highly integrated approaches involving liver-stage research, "omics" studies, and cutting-edge genetic editing technologies, combined with in vitro culture systems or unique animal models, are needed to accelerate the discovery of candidates for a late-arresting, genetically attenuated parasite vaccine.


Subject(s)
Liver/immunology , Malaria Vaccines/genetics , Malaria, Falciparum/prevention & control , Malaria, Vivax/prevention & control , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Sporozoites/immunology , Animals , Disease Models, Animal , Gamma Rays , Genetic Engineering/methods , Humans , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Mice , Plasmodium berghei/chemistry , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/radiation effects , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/radiation effects , Plasmodium vivax/chemistry , Plasmodium vivax/genetics , Plasmodium vivax/radiation effects , Plasmodium yoelii/chemistry , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Plasmodium yoelii/radiation effects , Sporozoites/chemistry , Sporozoites/genetics , Sporozoites/radiation effects , Vaccines, Attenuated
8.
Sci Rep ; 7(1): 11991, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28931852

ABSTRACT

Pichia pastoris is a simple and powerful expression platform that has the ability to produce a wide variety of recombinant proteins, ranging from simple peptides to complex membrane proteins. A well-established fermentation strategy is available comprising three main phases: a batch phase, followed by a glycerol fed-batch phase that increases cell density, and finally an induction phase for product expression using methanol as the inducer. We previously used this three-phase strategy at the 15-L scale to express three different AMA1-DiCo-based malaria vaccine candidates to develop a vaccine cocktail. For two candidates, we switched to a two-phase strategy lacking the intermediate glycerol fed-batch phase. The new strategy not only provided a more convenient process flow but also achieved 1.5-fold and 2.5-fold higher space-time yields for the two candidates, respectively, and simultaneously reduced the final cell mass by a factor of 1.3, thus simplifying solid-liquid separation. This strategy also reduced the quantity of host cell proteins that remained to be separated from the two vaccine candidates (by 34% and 13%, respectively), thus reducing the effort required in the subsequent purification steps. Taken together, our new fermentation strategy increased the overall fermentation performance for the production of two different AMA1-DiCo-based vaccine candidates.


Subject(s)
Antigens, Protozoan/metabolism , Biotechnology/methods , Malaria Vaccines/metabolism , Membrane Proteins/metabolism , Pichia/metabolism , Protozoan Proteins/metabolism , Technology, Pharmaceutical/methods , Antigens, Protozoan/genetics , Fermentation , Malaria Vaccines/genetics , Membrane Proteins/genetics , Pichia/genetics , Protozoan Proteins/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/metabolism
9.
J Biol Chem ; 292(28): 11960-11969, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28533429

ABSTRACT

FREP1 in mosquito midguts facilitates Plasmodium falciparum parasite transmission. The fibrinogen-like (FBG) domain of FREP1 is highly conserved (>90% identical) among Anopheles species from different continents, suggesting that anti-FBG antibodies may block malaria transmission to all anopheline mosquitoes. Using standard membrane-feeding assays, anti-FREP1 polyclonal antibodies significantly blocked transmission of Plasmodium berghei and Plasmodium vivax to Anopheles gambiae and Anopheles dirus, respectively. Furthermore, in vivo studies of mice immunized with FBG achieved >75% blocking efficacy of P. berghei to A. gambiae without triggering immunopathology. Anti-FBG serum also reduced >81% of P. falciparum infection to A. gambiae Finally, we showed that FBG interacts with Plasmodium gametocytes and ookinetes, revealing the molecular mechanism of its antibody transmission-blocking activity. Collectively, our data support that FREP1-mediated Plasmodium transmission to mosquitoes is a conserved pathway and that targeting the FBG domain of FREP1 will limit the transmission of multiple Plasmodium species to multiple Anopheles species.


Subject(s)
Anopheles/metabolism , Insect Proteins/therapeutic use , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Amino Acid Sequence , Animals , Anopheles/immunology , Anopheles/parasitology , Antibodies, Blocking/analysis , Conserved Sequence , Female , Germ Cells/immunology , Germ Cells/metabolism , Humans , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Proteins/metabolism , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Malaria, Vivax/blood , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Male , Mice , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Plasmodium berghei/growth & development , Plasmodium berghei/immunology , Plasmodium falciparum/growth & development , Plasmodium vivax/growth & development , Plasmodium vivax/immunology , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/metabolism , Vaccines, Synthetic/therapeutic use
10.
Mikrobiyol Bul ; 51(1): 41-51, 2017 Jan.
Article in Turkish | MEDLINE | ID: mdl-28283009

ABSTRACT

Malaria is caused by the protozoan parasite Plasmodium, the leading cause of death amongst the parasitic diseases. The disease is transmitted to human by the bites of female Anopheles mosquitoes. According to the World Health Organization (WHO) data, there were an estimated 214 million malaria cases and estimated 438.000 deaths occurred worldwide, in 2015. It is observed that 90% of all the deaths due to malaria occur in Africa. 78% of these cases were children who are under five years old. Intensive malaria interventions helped to reduce malaria incidence by 37% between 2000 and 2015. Malaria is a curable disease if diagnosed and treated promptly and correctly. Drug resistance has developed against almost all anti-malarial drugs and an effective vaccine against malaria has not been developed yet. Vaccine studies initiated 40 years ago by sterile immunity against falciparum malaria through immunization by exposure to 1000 irradiated mosquitoes. Complex structures, complicated life cycles and various antigenic structures of Plasmodium species make vaccination studies difficult. Circumsporozoite protein (CSP), the most extensively studied protein is also present in the content of the vaccine candidate RTS,S which is currently closest to get license. CSP was the first described Plasmodium antigen because of its important role during initiation of the parasitic infection. CSP is the major surface coat protein of Plasmodium parasite. CSP is a soluble protein and recombinant form of the CSP can be produced in Escherichia coli. NANP repeat region is a target site for host antibodies. Recently many DNA, RNA and protein vaccine candidates are being developed against malaria. According to WHO, in the next 20 years period, malaria vaccine can be developed. In this study we aimed to produce recombinant CSP (rCSP). Initially, P.falciparum CSP gene was amplified by PCR. CSP gene was cloned in to the pJET cloning vector. The gene subcloned to the pET100 protein expression vector. E.coli cells were used for protein expression. After this process, purification and endotoxin removal protocols were performed. As a result, 1182 bp CSP gene was obtained from P.falciparum genomic DNA. Accuracy of cloning and DNA sequence of the CSP gene was determined with DNA sequence analysis. The gene sequence was recorded to the GenBank with a registration no KT315396. rCSP was expressed in E.coli cells. The existence of rCSP was verifiedwith Western Blot method and was purified and removed from endotoxins. rCSP aminoacid sequence and 3D shape was obtained.We believe that the production of recombinant CSP will enable us to contribute to the further malaria vaccine studies in our laboratory and country.


Subject(s)
Epitopes , Malaria Vaccines , Malaria/parasitology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Anopheles/parasitology , Child, Preschool , DNA, Protozoan/chemistry , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Female , Humans , Infant , Malaria/epidemiology , Malaria/prevention & control , Malaria/transmission , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria Vaccines/metabolism , Mosquito Vectors/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/metabolism
11.
Elife ; 62017 02 14.
Article in English | MEDLINE | ID: mdl-28195038

ABSTRACT

Invasion of erythrocytes by Plasmodial merozoites is a composite process involving the interplay of several proteins. Among them, the Plasmodium falciparum Cysteine-Rich Protective Antigen (PfCyRPA) is a crucial component of a ternary complex, including Reticulocyte binding-like Homologous protein 5 (PfRH5) and the RH5-interacting protein (PfRipr), essential for erythrocyte invasion. Here, we present the crystal structures of PfCyRPA and its complex with the antigen-binding fragment of a parasite growth inhibitory antibody. PfCyRPA adopts a 6-bladed ß-propeller structure with similarity to the classic sialidase fold, but it has no sialidase activity and fulfills a purely non-enzymatic function. Characterization of the epitope recognized by protective antibodies may facilitate design of peptidomimetics to focus vaccine responses on protective epitopes. Both in vitro and in vivo anti-PfCyRPA and anti-PfRH5 antibodies showed more potent parasite growth inhibitory activity in combination than on their own, supporting a combined delivery of PfCyRPA and PfRH5 in vaccines.


Subject(s)
Antibodies, Protozoan/chemistry , Antibodies, Protozoan/metabolism , Antigens, Protozoan/chemistry , Antigens, Protozoan/metabolism , Malaria Vaccines/chemistry , Malaria Vaccines/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Conformation
12.
Elife ; 62017 02 14.
Article in English | MEDLINE | ID: mdl-28195530

ABSTRACT

Plasmodium falciparum causes malaria in humans with over 450,000 deaths annually. The asexual blood stage involves invasion of erythrocytes by merozoites, in which they grow and divide to release daughter merozoites, which in turn invade new erythrocytes perpetuating the cycle responsible for malaria. A key step in merozoite invasion is the essential binding of PfRh5/CyRPA/PfRipr complex to basigin, a step linked to the formation of a pore between merozoites and erythrocytes. We show CyRPA interacts directly with PfRh5. An invasion inhibitory monoclonal antibody to CyRPA blocks binding of CyRPA to PfRh5 and complex formation thus illuminating the molecular mechanism for inhibition of parasite growth. We determined the crystal structures of CyRPA alone and in complex with an antibody Fab fragment. CyRPA has a six-bladed ß-propeller fold, and we identify the region that interacts with PfRh5. This functionally conserved epitope is a potential target for vaccines against P. falciparum.


Subject(s)
Antibodies, Protozoan/chemistry , Antibodies, Protozoan/metabolism , Antigens, Protozoan/chemistry , Antigens, Protozoan/metabolism , Carrier Proteins/metabolism , Malaria Vaccines/chemistry , Malaria Vaccines/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Conformation
13.
PLoS One ; 11(12): e0166814, 2016.
Article in English | MEDLINE | ID: mdl-27911910

ABSTRACT

Experimental immunization with radiation attenuated sporozoites (RAS) and genetically attenuated sporozoites has proved to be a promising approach for malaria vaccine development. However, parasite biomarkers of growth attenuation and enhanced immune protection in response to radiation remain poorly understood. Here, we report on the effect of an attenuating dose of γ-irradiation (15 krad) on the Plasmodium falciparum sporozoite (PfSPZ) ultrastructure by electron microscopy, growth rate of liver stage P. falciparum in liver cell cultures, and genome-wide transcriptional profile of liver stage parasites by microarray. We find that γ-irradiation treated PfSPZ retained a normal cellular structure except that they were vacuous with a partially disrupted plasma membrane and inner membrane complex. A similar infection rate was observed by γ-irradiation-treated and untreated PfSPZ in human HCO-4 liver cells (0.47% versus 0.49%, respectively) on day 3 post-infection. In the microarray studies, cumulatively, 180 liver stage parasite genes were significantly transcriptionally altered on day 3 and/or 6 post-infection. Among the transcriptionally altered biomarkers, we identified a signature of seven candidate parasite genes that associated with functionally diverse pathways that may regulate radiation induced cell cycle arrest of the parasite within the hepatocyte. A repertoire of 14 genes associated with protein translation is transcriptionally overexpressed within the parasite by radiation. Additionally, 37 genes encode proteins expressed on the cell surface or exported into the host cell, 4 encode membrane associated transporters, and 10 encode proteins related to misfolding and stress-related protein processing. These results have significantly increased the repertoire of novel targets for 1) biomarkers of safety to define proper attenuation, 2) generating genetically attenuated parasite vaccine candidates, and 3) subunit candidate vaccines against liver stage malaria.


Subject(s)
Gamma Rays , Gene Expression Regulation/radiation effects , Liver/metabolism , Malaria Vaccines/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Sporozoites/metabolism , Animals , Anopheles , Biomarkers/metabolism , Cell Line , Female , Humans , Liver/parasitology , Malaria, Falciparum/metabolism , Malaria, Falciparum/prevention & control , Vaccines, Attenuated/metabolism
14.
Comput Biol Med ; 76: 24-9, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27393958

ABSTRACT

BACKGROUND: Recombinant protein technology has revolutionized the world of biology and medicine. Following this progress, fusion protein technology, as a novel innovation, has opened new horizons for the development of proteins that do not naturally exist. Fusion proteins are generated via genetically fusing two or more genes coding for separate proteins, thus the product is a single protein having functional properties of both proteins. As an indispensable element in fusion protein construction, linkers are used to separate the functional domains in order to improve their expression, folding and stability. METHOD: We computationally fused an antigen and an adjuvant together using different linkers to obtain a two-domain fusion construct which can potentially act as an oral vaccine candidate against malaria. We then predicted the structures computationally to find out the probable folding of each domain in the designed construct. RESULTS: One of the fusion constructs was selected based on the highest value for C-score. Ramchandran Plot analysis represented that most residues were fallen in favorable regions. CONCLUSION: Our in silico analysis showed that (GGGGS)3 linker confers the best structure and stability for our target fusion protein.


Subject(s)
Malaria Vaccines/chemistry , Protozoan Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Vaccines, Synthetic/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/metabolism , Models, Molecular , Protein Conformation , Protein Folding , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vaccines, Synthetic/genetics , Vaccines, Synthetic/metabolism
15.
Biotechnol Bioeng ; 112(7): 1297-305, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25615702

ABSTRACT

Malaria is a vector-borne disease affecting more than two million people and accounting for more than 600,000 deaths each year, especially in developing countries. The most serious form of malaria is caused by Plasmodium falciparum. The complex life cycle of this parasite, involving pre-erythrocytic, asexual and sexual stages, makes vaccine development cumbersome but also offers a broad spectrum of vaccine candidates targeting exactly those stages. Vaccines targeting the sexual stage of P. falciparum are called transmission-blocking vaccines (TBVs). They do not confer protection for the vaccinated individual but aim to reduce or prevent the transmission of the parasite within a population and are therefore regarded as an essential tool in the fight against the disease. Malaria predominantly affects large populations in developing countries, so TBVs need to be produced in large quantities at low cost. Combining the advantages of eukaryotic expression with a virtually unlimited upscaling potential and a good product safety profile, plant-based expression systems represent a suitable alternative for the production of TBVs. We report here the high level (300 µg/g fresh leaf weight (FLW)) transient expression in Nicotiana benthamiana leaves of an effective TBV candidate based on a fusion protein F0 comprising Pfs25 and the C0-domain of Pfs230, and the implementation of a simple and cost-effective heat treatment step for purification that yields intact recombinant protein at >90% purity with a recovery rate of >70%. The immunization of mice clearly showed that antibodies raised against plant-derived F0 completely blocked the formation of oocysts in a malaria transmission-blocking assay (TBA) making F0 an interesting TBV candidate or a component of a multi-stage malaria vaccine cocktail.


Subject(s)
Antigens, Protozoan/isolation & purification , Fractional Precipitation , Malaria Vaccines/isolation & purification , Nicotiana/metabolism , Plants, Genetically Modified/metabolism , Protozoan Proteins/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Hot Temperature , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria Vaccines/metabolism , Mice , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Nicotiana/genetics , Vaccination/methods
16.
Biotechnol J ; 8(6): 738-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23447434

ABSTRACT

The identification of optimal expression conditions for state-of-the-art production of pharmaceutical proteins is a very time-consuming and expensive process. In this report a method for rapid and reproducible optimization of protein expression in an in-house designed small-scale BIOSTAT® multi-bioreactor plant is described. A newly developed BioPAT® MFCS/win Design of Experiments (DoE) module (Sartorius Stedim Systems, Germany) connects the process control system MFCS/win and the DoE software MODDE® (Umetrics AB, Sweden) and enables therefore the implementation of fully automated optimization procedures. As a proof of concept, a commercial Pichia pastoris strain KM71H has been transformed for the expression of potential malaria vaccines. This approach has allowed a doubling of intact protein secretion productivity due to the DoE optimization procedure compared to initial cultivation results. In a next step, robustness regarding the sensitivity to process parameter variability has been proven around the determined optimum. Thereby, a pharmaceutical production process that is significantly improved within seven 24-hour cultivation cycles was established. Specifically, regarding the regulatory demands pointed out in the process analytical technology (PAT) initiative of the United States Food and Drug Administration (FDA), the combination of a highly instrumented, fully automated multi-bioreactor platform with proper cultivation strategies and extended DoE software solutions opens up promising benefits and opportunities for pharmaceutical protein production.


Subject(s)
Bioreactors/microbiology , Biotechnology , Recombinant Proteins/biosynthesis , Research Design , Biotechnology/instrumentation , Biotechnology/methods , Industrial Microbiology , Malaria Vaccines/metabolism , Pichia/metabolism , Protozoan Proteins/metabolism
17.
Bioengineered ; 4(3): 164-7, 2013.
Article in English | MEDLINE | ID: mdl-23090388

ABSTRACT

Malaria is an infectious disease that threatens half of the world's population. This debilitating disease is caused by infection from parasites of the genus Plasmodium. Insecticides, bed nets and drug therapies have lowered the prevalence and death rate associated with malaria but this disease continues to plague many populations around the world. In recent years, many organizations have suggested developing methods for a complete eradication of malaria. The most straightforward and effective method for this potential eradication will be through the development of a low-cost vaccine. To achieve eradication, it will be necessary to develop new vaccine candidates and novel systems for both the production and delivery of these vaccines. Recently, the green algae Chlamydomonas reinhardtii has been used for the recombinant expression of malaria vaccine candidates including the transmission blocking vaccine candidate Pfs48/45. Here, we discuss the potential of this research on the future development of a low-cost malaria vaccine candidate.


Subject(s)
Chlamydomonas reinhardtii/genetics , Gene Expression , Malaria Vaccines/genetics , Malaria Vaccines/metabolism , Malaria/prevention & control , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Chlamydomonas reinhardtii/metabolism , Humans , Malaria/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Plasmodium falciparum/physiology
18.
Trends Parasitol ; 28(1): 23-30, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22178537

ABSTRACT

The global agenda for malaria eradication would benefit from development of a highly efficacious vaccine that protects against disease and interrupts transmission of Plasmodium falciparum. It is likely that such a vaccine will be multi-component, with antigens from different stages of the parasite life cycle. In this review, inclusion of blood stage antigens in such a vaccine is discussed. Erythrocyte binding-like (EBL) and P. falciparum reticulocyte binding-like (PfRh) proteins are reviewed with respect to their function in erythrocyte invasion, their role in eliciting antibodies contributing to protective immunity and reduction of invasion, leading subsequently to inhibition of parasite multiplication.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/parasitology , Humans , Life Cycle Stages/immunology , Malaria Vaccines/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Reticulocytes/immunology , Reticulocytes/metabolism
19.
Mem Inst Oswaldo Cruz ; 106 Suppl 1: 167-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21881771

ABSTRACT

Recently, we described the improved immunogenicity of new malaria vaccine candidates based on the expression of fusion proteins containing immunodominant epitopes of merozoites and Salmonella enterica serovar Typhimurium flagellin (FliC) protein as an innate immune agonist. Here, we tested whether a similar strategy, based on an immunodominant B-cell epitope from malaria sporozoites, could also generate immunogenic fusion polypeptides. A recombinant His6-tagged FliC protein containing the C-terminal repeat regions of the VK210 variant of Plasmodium vivax circumsporozoite (CS) protein was constructed. This recombinant protein was successfully expressed in Escherichia coli as soluble protein and was purified by affinity to Ni-agarose beads followed by ion exchange chromatography. A monoclonal antibody specific for the CS protein of P. vivax sporozoites (VK210) was able to recognise the purified protein. C57BL/6 mice subcutaneously immunised with the recombinant fusion protein in the absence of any conventional adjuvant developed protein-specific systemic antibody responses. However, in mice genetically deficient in expression of TLR5, this immune response was extremely low. These results extend our previous observations concerning the immunogenicity of these recombinant fusion proteins and provide evidence that the main mechanism responsible for this immune activation involves interactions with TLR5, which has not previously been demonstrated for any recombinant FliC fusion protein.


Subject(s)
Flagellin/immunology , Immunodominant Epitopes/immunology , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium falciparum/immunology , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/immunology , Animals , Antibodies, Protozoan/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Escherichia coli Proteins/immunology , Flagellin/metabolism , Immunodominant Epitopes/metabolism , Malaria Vaccines/metabolism , Malaria, Vivax/immunology , Mice , Mice, Inbred C57BL , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Salmonella typhimurium/metabolism , Toll-Like Receptor 5/immunology
20.
Proc Natl Acad Sci U S A ; 108(18): 7553-8, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502513

ABSTRACT

The ligands that pathogens use to invade their target cells have often proven to be good targets for vaccine development. However, Plasmodium falciparum has redundant ligands that mediate invasion of erythrocytes. The first requirement for the development of a successful ligand-blocking malaria vaccine is the demonstration that antibodies induced to each ligand can block the erythrocyte invasion of parasites with polymorphic sequences. Because of P. falciparum's redundancy in erythrocyte invasion, each ligand needs to be studied under artificial conditions in which parasite invasion is restricted in its use of alternative pathways. Here we investigate the role of erythrocyte-binding antigen 175 (EBA-175), a parasite ligand that binds to sialic acid on glycophorin A, in the invasion of erythrocytes by 10 P. falciparum clones under conditions in which invasion is partially limited to the EBA-175-glycophorin A pathway, using chymotrypsin-treated erythrocytes. We show that the ability to invade erythrocytes for both sialic acid-independent and sialic acid-dependent pathways requires the EBA-175-glycophorin A pathway for erythrocyte invasion. Importantly, antibodies against region II of EBA-175 from the 3D7 clone blocked invasion of chymotrypsin-treated erythrocytes by >50% by all parasite clones studied, including those with multiple different mutations described in the literature. The one exception was FCR3, which had a similar sequence to 3D7 but only 30% inhibition of invasion of chymotrypsin-treated erythrocytes, indicating alternative pathways for invasion of chymotrypsin-treated erythrocytes. Our findings suggest that antibodies to region II of EBA-175, as one component of a ligand-blocking malaria vaccine, are largely unaffected by polymorphism in EBA-175.


Subject(s)
Antibodies, Protozoan/pharmacology , Antigens, Protozoan/immunology , Erythrocytes/parasitology , Glycophorins/metabolism , Host-Parasite Interactions/drug effects , Malaria Vaccines/immunology , Malaria/prevention & control , Protozoan Proteins/immunology , Antigens, Protozoan/metabolism , Chymotrypsin , Erythrocytes/metabolism , Host-Parasite Interactions/immunology , Humans , Immunoglobulin G/metabolism , Malaria Vaccines/metabolism , Protozoan Proteins/metabolism , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...