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










Publication year range
1.
Sci Rep ; 8(1): 1118, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348479

ABSTRACT

Vaccine development against Plasmodium vivax malaria lags behind that for Plasmodium falciparum. To narrow this gap, we administered recombinant antigens based on P. vivax circumsporozoite protein (CSP) to mice. We expressed in Pichia pastoris two chimeric proteins by merging the three central repeat regions of different CSP alleles (VK210, VK247, and P. vivax-like). The first construct (yPvCSP-AllFL) contained the fused repeat regions flanked by N- and C-terminal regions. The second construct (yPvCSP-AllCT) contained the fused repeat regions and the C-terminal domain, plus RI region. Mice were vaccinated with three doses of yPvCSP in adjuvants Poly (I:C) or Montanide ISA720. We also used replication-defective adenovirus vectors expressing CSP of human serotype 5 (AdHu5) and chimpanzee serotype 68 (AdC68) for priming mice which were subsequently boosted twice with yPvCSP proteins in Poly (I:C) adjuvant. Regardless of the regime used, immunized mice generated high IgG titres specific to all CSP alleles. After challenge with P. berghei ANKA transgenic parasites expressing Pb/PvVK210 or Pb/PvVK247 sporozoites, significant time delays for parasitemia were observed in all vaccinated mice. These vaccine formulations should be clinically tried for their potential as protective universal vaccine against P. vivax malaria.


Subject(s)
Malaria Vaccines/immunology , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Recombinant Proteins/immunology , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Antibody Affinity/immunology , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , Genetic Vectors/chemistry , Immunization , Immunogenicity, Vaccine , Immunoglobulin G/blood , Immunoglobulin G/immunology , Malaria Vaccines/genetics , Malaria, Vivax/mortality , Mice , Plasmodium vivax/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
2.
Cell Host Microbe ; 22(6): 766-776.e4, 2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29241041

ABSTRACT

Artemisinin and its derivatives (ARTs) are frontline antimalarial drugs. However, ART monotherapy is associated with a high frequency of recrudescent infection, resulting in treatment failure. A subset of parasites is thought to undergo ART-induced latency, but the mechanisms remain unknown. Here, we report that ART treatment results in phosphorylation of the parasite eukaryotic initiation factor-2α (eIF2α), leading to repression of general translation and latency induction. Enhanced phosphorylated eIF2α correlates with high rates of recrudescence following ART, and inhibiting eIF2α dephosphorylation renders parasites less sensitive to ART treatment. ART-induced eIF2α phosphorylation is mediated by the Plasmodium eIF2α kinase, PK4. Overexpression of a PK4 dominant-negative or pharmacological inhibition of PK4 blocks parasites from entering latency and abolishes recrudescence after ART treatment of infected mice. These results show that translational control underlies ART-induced latency and that interference with this stress response may resolve the clinical problem of recrudescent infection.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Plasmodium/drug effects , Plasmodium/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Mice , Phosphorylation , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational
3.
Front Immunol ; 8: 1275, 2017.
Article in English | MEDLINE | ID: mdl-29075260

ABSTRACT

Plasmodium vivax is the most common species that cause malaria outside of the African continent. The development of an efficacious vaccine would contribute greatly to control malaria. Recently, using bacterial and adenoviral recombinant proteins based on the P. vivax circumsporozoite protein (CSP), we demonstrated the possibility of eliciting strong antibody-mediated immune responses to each of the three allelic forms of P. vivax CSP (PvCSP). In the present study, recombinant proteins representing the PvCSP alleles (VK210, VK247, and P. vivax-like), as well as a hybrid polypeptide, named PvCSP-All epitopes, were generated. This hybrid containing the conserved C-terminal of the PvCSP and the three variant repeat domains in tandem were successfully produced in the yeast Pichia pastoris. After purification and biochemical characterization, they were used for the experimental immunization of C57BL/6 mice in a vaccine formulation containing the adjuvant Poly(I:C). Immunization with a recombinant protein expressing all three different allelic forms in fusion elicited high IgG antibody titers reacting with all three different allelic variants of PvCSP. The antibodies targeted both the C-terminal and repeat domains of PvCSP and recognized the native protein on the surface of P. vivax sporozoites. More importantly, mice that received the vaccine formulation were protected after challenge with chimeric Plasmodium berghei sporozoites expressing CSP repeats of P. vivax sporozoites (Pb/PvVK210). Our results suggest that it is possible to elicit protective immunity against one of the most common PvCSP alleles using soluble recombinant proteins expressed by P. pastoris. These recombinant proteins are promising candidates for clinical trials aiming to develop a multiallele vaccine against P. vivax malaria.

4.
Vaccine ; 34(38): 4501-4506, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27502569

ABSTRACT

A number of studies have shown that CD8+ T cells mediate protective anti-malaria immunity in a mouse model. However, whether human CD8+ T cells play a role in protection against malaria remains unknown. We recently established human immune system (HIS) mice harboring functional human CD8+ T cells (HIS-CD8 mice) by transduction with HLA-A∗0201 and certain human cytokines using recombinant adeno-associated virus-based gene transfer technologies. These HIS-CD8 mice mount a potent, antigen-specific HLA-A∗0201-restricted human CD8+ T-cell response upon immunization with a recombinant adenovirus expressing a human malaria antigen, the Plasmodium falciparum circumsporozoite protein (PfCSP), termed AdPfCSP. In the present study, we challenged AdPfCSP-immunized HIS-CD8 mice with transgenic Plasmodium berghei sporozoites expressing full-length PfCSP and found that AdPfCSP-immunized (but not naïve) mice were protected against subsequent malaria challenge. The level of the HLA-A∗0201-restricted, PfCSP-specific human CD8+ T-cell response was closely correlated with the level of malaria protection. Furthermore, depletion of human CD8+ T cells from AdPfCSP-immunized HIS-CD8 mice almost completely abolished the anti-malaria immune response. Taken together, our data show that human CD8+ T cells mediate protective anti-malaria immunity in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Animals , Dependovirus , Genetic Vectors , HLA-A2 Antigen/genetics , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Plasmodium falciparum , Protozoan Proteins/immunology , Sporozoites
5.
PLoS Pathog ; 12(1): e1005370, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26735921

ABSTRACT

Plasmodium salivary sporozoites are the infectious form of the malaria parasite and are dormant inside salivary glands of Anopheles mosquitoes. During dormancy, protein translation is inhibited by the kinase UIS1 that phosphorylates serine 59 in the eukaryotic initiation factor 2α (eIF2α). De-phosphorylation of eIF2α-P is required for the transformation of sporozoites into the liver stage. In mammalian cells, the de-phosphorylation of eIF2α-P is mediated by the protein phosphatase 1 (PP1). Using a series of genetically knockout parasites we showed that in malaria sporozoites, contrary to mammalian cells, the eIF2α-P phosphatase is a member of the PP2C/PPM phosphatase family termed UIS2. We found that eIF2α was highly phosphorylated in uis2 conditional knockout sporozoites. These mutant sporozoites maintained the crescent shape after delivery into mammalian host and lost their infectivity. Both uis1 and uis2 were highly transcribed in the salivary gland sporozoites but uis2 expression was inhibited by the Pumilio protein Puf2. The repression of uis2 expression was alleviated when sporozoites developed into liver stage. While most eukaryotic phosphatases interact transiently with their substrates, UIS2 stably bound to phosphorylated eIF2α, raising the possibility that high-throughput searches may identify chemicals that disrupt this interaction and prevent malaria infection.


Subject(s)
Host-Parasite Interactions/physiology , Malaria/parasitology , Phosphoric Monoester Hydrolases/metabolism , Plasmodium berghei/enzymology , Plasmodium berghei/growth & development , Sporozoites/enzymology , Sporozoites/growth & development , Animals , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Gene Knockout Techniques , Humans , Immunoblotting , Immunoprecipitation , Life Cycle Stages , Mice , Phosphorylation
6.
J Immunol Methods ; 427: 42-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410104

ABSTRACT

In this study, we developed human immune system (HIS) mice that possess functional human CD4+ T cells and B cells, named HIS-CD4/B mice. HIS-CD4/B mice were generated by first introducing HLA class II genes, including DR1 and DR4, along with genes encoding various human cytokines and human B cell activation factor (BAFF) to NSG mice by adeno-associated virus serotype 9 (AAV9) vectors, followed by engrafting human hematopoietic stem cells (HSCs). HIS-CD4/B mice, in which the reconstitution of human CD4+ T and B cells resembles to that of humans, produced a significant level of human IgG against Plasmodium falciparum circumsporozoite (PfCS) protein upon immunization. CD4+ T cells in HIS-CD4/B mice, which possess central and effector memory phenotypes like those in humans, are functional, since PfCS protein-specific human CD4+ T cells secreting IFN-γ and IL-2 were detected in immunized HIS-CD4/B mice. Lastly, PfCS protein-immunized HIS-CD4/B mice were protected from in vivo challenge with transgenic P. berghei sporozoites expressing the PfCS protein. The immune sera collected from protected HIS-CD4/B mice reacted against transgenic P. berghei sporozoites expressing the PfCS protein and also inhibited the parasite invasion into hepatocytes in vitro. Taken together, these studies show that our HIS-CD4/B mice could mount protective human anti-malaria immunity, consisting of human IgG and human CD4+ T cell responses both specific for a human malaria antigen.


Subject(s)
Disease Models, Animal , Immunity, Humoral/immunology , Malaria, Falciparum/immunology , Mice, Transgenic/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Heterografts , Histocompatibility Antigens Class II , Humans , Malaria Vaccines , Mice , Protozoan Proteins/immunology
7.
Malar J ; 14: 324, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26289816

ABSTRACT

BACKGROUND: Emerging resistance of the malaria parasite Plasmodium to current therapies underscores the critical importance of exploring novel strategies for disease eradication. Plasmodium species are obligate intracellular protozoan parasites. They rely on an unusual form of substrate-dependent motility for their migration on and across host-cell membranes and for host cell invasion. This peculiar motility mechanism is driven by the 'glideosome', an actin-myosin associated, macromolecular complex anchored to the inner membrane complex of the parasite. Myosin A, actin, aldolase, and thrombospondin-related anonymous protein (TRAP) constitute the molecular core of the glideosome in the sporozoite, the mosquito stage that brings the infection into mammals. METHODS: Virtual library screening of a large compound library against the PfAldolase-TRAP complex was used to identify candidate compounds that stabilize and prevent the disassembly of the glideosome. The mechanism of these compounds was confirmed by biochemical, biophysical and parasitological methods. RESULTS: A novel inhibitory effect on the parasite was achieved by stabilizing a protein-protein interaction within the glideosome components. Compound 24 disrupts the gliding and invasive capabilities of Plasmodium parasites in in vitro parasite assays. A high-resolution, ternary X-ray crystal structure of PfAldolase-TRAP in complex with compound 24 confirms the mode of interaction and serves as a platform for future ligand optimization. CONCLUSION: This proof-of-concept study presents a novel approach to anti-malarial drug discovery and design. By strengthening a protein-protein interaction within the parasite, an avenue towards inhibiting a previously "undruggable" target is revealed and the motility motor responsible for successful invasion of host cells is rendered inactive. This study provides new insights into the malaria parasite cell invasion machinery and convincingly demonstrates that liver cell invasion is dramatically reduced by 95 % in the presence of the small molecule stabilizer compound 24.


Subject(s)
Fructose-Bisphosphate Aldolase/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/chemistry , Protozoan Proteins/metabolism , Animals , Cell Line , Cell Survival/drug effects , Crystallography, X-Ray , Fructose-Bisphosphate Aldolase/chemistry , Hepatocytes/drug effects , Humans , Membrane Proteins/chemistry , Molecular Docking Simulation , Multiprotein Complexes/drug effects , Plasmodium falciparum/chemistry , Protein Stability/drug effects , Protozoan Proteins/chemistry , Rabbits , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/toxicity , Surface Plasmon Resonance
8.
Infect Immun ; 82(2): 793-807, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478093

ABSTRACT

Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.


Subject(s)
Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Female , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Vivax/immunology , Mice , Mice, Inbred C57BL , Plasmodium vivax/genetics , Poly I-C/administration & dosage , Protozoan Proteins/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
9.
Eukaryot Cell ; 12(2): 161-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23243065

ABSTRACT

The life cycles of apicomplexan parasites such as Plasmodium spp. and Toxoplasma gondii are complex, consisting of proliferative and latent stages within multiple hosts. Dramatic transformations take place during the cycles, and they demand precise control of gene expression at all levels, including translation. This review focuses on the mechanisms that regulate translational control in Plasmodium and Toxoplasma, with a particular emphasis on the phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eIF2α). Phosphorylation of eIF2α (eIF2α∼P) is a conserved mechanism that eukaryotic cells use to repress global protein synthesis while enhancing gene-specific translation of a subset of mRNAs. Elevated levels of eIF2α∼P have been observed during latent stages in both Toxoplasma and Plasmodium, indicating that translational control plays a role in maintaining dormancy. Parasite-specific eIF2α kinases and phosphatases are also required for proper developmental transitions and adaptation to cellular stresses encountered during the life cycle. Identification of small-molecule inhibitors of apicomplexan eIF2α kinases may selectively interfere with parasite translational control and lead to the development of new therapies to treat malaria and toxoplasmosis.


Subject(s)
Plasmodium/genetics , Protein Biosynthesis , Toxoplasma/genetics , Animals , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Host-Parasite Interactions , Humans , Malaria/parasitology , Phosphorylation , Plasmodium/metabolism , Plasmodium/physiology , Protein Processing, Post-Translational , Protozoan Proteins/metabolism , Sporozoites/physiology , Toxoplasma/metabolism , Toxoplasma/physiology , Toxoplasmosis/parasitology
10.
Biomaterials ; 33(27): 6476-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22695068

ABSTRACT

Biomaterials that modulate innate and adaptive immune responses are receiving increasing interest as adjuvants for eliciting protective immunity against a variety of diseases. Previous results have indicated that self-assembling ß-sheet peptides, when fused with short peptide epitopes, can act as effective adjuvants and elicit robust and long-lived antibody responses. Here we investigated the mechanism of immunogenicity and the quality of antibody responses raised by a peptide epitope from Plasmodium falciparum circumsporozoite (CS) protein, (NANP)(3),conjugated to the self-assembling peptide domain Q11. The mechanism of adjuvant action was investigated in knockout mice with impaired MyD88, NALP3, TLR-2, or TLR-5 function, and the quality of antibodies raised against (NANP)(3)-Q11 was assessed using a transgenic sporozoite neutralizing (TSN) assay for malaria infection. (NANP)(3)-Q11 self-assembled into nanofibers, and antibody responses lasted up to 40 weeks in C57BL/6 mice. The antibody responses were T cell- and MyD88-dependent. Sera from mice primed with either irradiated sporozoites or a synthetic peptide, (T1BT*)(4)-P3C, and boosted with (NANP)(3)-Q11 showed significant increases in antibody titers and significant inhibition of sporozoite infection in TSN assays. In addition, two different epitopes could be self-assembled together without compromising the strength or duration of the antibody responses raised against either of them, making these materials promising platforms for self-adjuvanting multi-antigenic immunotherapies.


Subject(s)
Antibody Formation/immunology , Epitopes/immunology , Malaria/immunology , Nanofibers/chemistry , Peptides/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myeloid Differentiation Factor 88/metabolism , Nanofibers/ultrastructure , Oligopeptides/chemistry , Oligopeptides/immunology , Peptides/chemistry , Protein Structure, Secondary , Sporozoites/immunology , T-Lymphocytes/immunology
11.
Proc Natl Acad Sci U S A ; 109(10): 3956-61, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22355110

ABSTRACT

In response to environmental stresses, the mammalian serine threonine kinases PERK, GCN2, HRI, and PKR phosphorylate the regulatory serine 51 of the eukaryotic translation initiation factor 2α (eIF2α) to inhibit global protein synthesis. Plasmodium, the protozoan that causes malaria, expresses three eIF2α kinases: IK1, IK2, and PK4. Like GCN2, IK1 regulates stress response to amino acid starvation. IK2 inhibits development of malaria sporozoites present in the mosquito salivary glands. Here we show that the phosphorylation by PK4 of the regulatory serine 59 of Plasmodium eIF2α is essential for the completion of the parasite's erythrocytic cycle that causes disease in humans. PK4 activity leads to the arrest of global protein synthesis in schizonts, where ontogeny of daughter merozoites takes place, and in gametocytes that infect Anopheles mosquitoes. The implication of these findings is that drugs that reduce PK4 activity should alleviate disease and inhibit malaria transmission.


Subject(s)
Plasmodium falciparum/metabolism , Protein Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism , Animals , Anopheles , Codon , DNA/genetics , Fungal Proteins/chemistry , Hep G2 Cells , Humans , Malaria/parasitology , Mice , Mice, Inbred C57BL , Models, Genetic , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Serine/chemistry
12.
J Immunol Methods ; 377(1-2): 47-52, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22306356

ABSTRACT

Plasmodium sporozoites are deposited in the skin of the mammalian host by Anopheles mosquitoes. To continue the life cycle, the sporozoites have to invade the host's hepatocytes, where they transform into exoerythrocytic forms (EEFs) inside a parasitophorous vacuole. During their route from the skin to the liver, the parasites traverse the capillary epithelium in the dermis to enter the blood circulation, and cross the endothelium of liver sinusoids to enter the parenchyma. Cell traversal by sporozoites is usually measured by quantifying dyes that enter or are released from cells during incubation with salivary gland sporozoites. These methods do not distinguish cell traversal from cell wounding. Here we validate an assay that quantifies cell traversal of sporozoites through monolayers of MDCK cells that form tight junctions. We compared cell traversal of wt sporozoites and of parasites lacking the Type I membrane protein TLP (TRAP-like protein) previously implicated in cell traversal. We provide direct evidence that TLP ko sporozoites are defective in cell traversal and that they are retained inside the MDCK cytoplasm. We then used the MDCK assay to study the effect of a monoclonal antibody (3D11) to the circumsporozoite protein (CSP) on the parasite's cell traversal. We show that 3D11 inhibits cell traversal at nanomolar concentrations. We conclude that antibodies elicited by CSP-based vaccines are likely to inhibit the migration of sporozoites from the skin to the liver.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Animals , Cell Line , Dogs , Electric Impedance , Intracellular Signaling Peptides and Proteins/immunology , Tight Junctions/immunology
14.
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
15.
Vaccine ; 29(43): 7335-42, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21807053

ABSTRACT

Immunization of BALB/c mice with irradiated sporozoites (IrSp) of Plasmodium yoelii can lead to sterile immunity. The circumsporozoite protein (CSP) plays a dominant role in protection. Nevertheless after hyper-immunization with IrSp, complete protection is obtained in CSP-transgenic BALB/c mice that are T-cell tolerant to the CSP and cannot produce antibodies [CSP-Tg/JhT(-/-)]. This protection is mediated exclusively by CD8(+) T cells [1]. To identify the non-CSP protective T cell antigens, we studied the properties of 34 P. yoelii sporozoite antigens that are predicted to be secreted and to contain strong Kd-restricted CD8(+) T cell epitopes. The synthetic peptides corresponding to the epitopes were used to screen for the presence of peptide-specific CD8(+) T cells secreting interferon-γ (IFN-γ) in splenocytes from CSP-Tg/JhT(-/-) BALB/c mice hyper immunized with IrSp. However, the numbers of IFN-γ-secreting splenocytes specific for the non-CSP antigen-derived peptides were 20-100 times lower than those specific for the CSP-specific peptide. When mice were immunized with recombinant adenoviruses expressing selected non-CSP antigens, the animals were not protected against challenge with P. yoelii sporozoites although large numbers of CD8(+) specific T cells were generated.


Subject(s)
Antigens, Protozoan/immunology , Epitopes, T-Lymphocyte/immunology , Malaria Vaccines/immunology , Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Immunization , Interferon-gamma , Malaria/immunology , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Mice, Transgenic , Sporozoites/radiation effects
16.
Mem. Inst. Oswaldo Cruz ; 106(supl.1): 167-171, Aug. 2011. ilus, graf
Article in English | LILACS | ID: lil-597258

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)
Animals , Mice , Flagellin/immunology , Immunodominant Epitopes/immunology , Malaria Vaccines/immunology , Malaria, Vivax , Plasmodium falciparum/immunology , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/immunology , Antibodies, Protozoan/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte , Escherichia coli Proteins/immunology , Flagellin , Immunodominant Epitopes , Malaria Vaccines , Malaria, Vivax/immunology , Protozoan Proteins/immunology , Protozoan Proteins , Recombinant Fusion Proteins , Salmonella typhimurium , /immunology
17.
J Exp Med ; 207(7): 1465-74, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20584882

ABSTRACT

Sporozoites, the invasive form of malaria parasites transmitted by mosquitoes, are quiescent while in the insect salivary glands. Sporozoites only differentiate inside of the hepatocytes of the mammalian host. We show that sporozoite latency is an active process controlled by a eukaryotic initiation factor-2alpha (eIF2alpha) kinase (IK2) and a phosphatase. IK2 activity is dominant in salivary gland sporozoites, leading to an inhibition of translation and accumulation of stalled mRNAs into granules. When sporozoites are injected into the mammalian host, an eIF2alpha phosphatase removes the PO4 from eIF2alpha-P, and the repression of translation is alleviated to permit their transformation into liver stages. In IK2 knockout sporozoites, eIF2alpha is not phosphorylated and the parasites transform prematurely into liver stages and lose their infectivity. Thus, to complete their life cycle, Plasmodium sporozoites exploit the mechanism that regulates stress responses in eukaryotic cells.


Subject(s)
Culicidae/parasitology , Plasmodium berghei/enzymology , Salivary Glands/parasitology , Sporozoites/enzymology , eIF-2 Kinase/metabolism , Animals , Cell Line , Cytoplasmic Granules/metabolism , Gene Expression Regulation , Gene Targeting , Life Cycle Stages , Liver/metabolism , Liver/parasitology , Mice , Mice, Inbred C57BL , Phenotype , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Plasmodium berghei/cytology , Plasmodium berghei/pathogenicity , Plasmodium berghei/ultrastructure , Protein Biosynthesis , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Salivary Glands/cytology , Salivary Glands/ultrastructure , Sporozoites/cytology , Sporozoites/ultrastructure
18.
Antimicrob Agents Chemother ; 54(7): 2987-93, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20457823

ABSTRACT

Nitrogen-containing bisphosphonates, drugs used to treat bone resorption diseases, also have activity against a broad range of protists, including blood-stage Plasmodium spp. Here, we show that new-generation "lipophilic" bisphosphonates designed as anticancer agents that block protein prenylation also have potent activity against Plasmodium liver stages, with a high (>100) therapeutic index. Treatment of mice with the bisphosphonate BPH-715 and challenge with Plasmodium berghei sporozoites revealed complete protection (no blood-stage parasites after 28 days). There was also activity against blood-stage forms in vitro and a 4-day delay in the prepatent period in vivo. The lipophilic bisphosphonates have activity against a Plasmodium geranylgeranyl diphosphate synthase (GGPPS), as well as low nM activity against human farnesyl and geranylgeranyl diphosphate synthases. The most active inhibitor in vitro and in vivo had enzyme inhibitory activity similar to that of the other, less active compounds but was more lipophilic. Lipophilic bisphosphonates are thus promising leads for novel antimalarials that target liver-stage infection.


Subject(s)
Antimalarials/therapeutic use , Diphosphonates/therapeutic use , Liver/physiopathology , Plasmodium berghei/drug effects , Animals , Diphosphonates/chemistry , Hep G2 Cells , Humans , Mice , Models, Biological , Molecular Structure , Plasmodium berghei/growth & development , Plasmodium berghei/pathogenicity
19.
Proc Natl Acad Sci U S A ; 107(3): 1172-7, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20080619

ABSTRACT

There is yet no licensed vaccine against malaria, a serious human disease affecting mostly children, with an annual death rate of about one million. Plasmodia, the malaria-causing parasites, have two obligatory hosts: mammals or birds, in which they multiply asexually, and mosquitoes with sexual multiplication. The most common and serious type of malaria is caused by Plasmodium falciparum. The circumsporozoite protein (CSP), a major surface antigen of sporozoites, is a protective antigen. A unique feature of P. falciparum CSP is its large central domain composed of over 30 tetrapeptide repeats of Asn-Ala-Asn-Pro (NANP). Several NANP peptide-protein conjugates were tested clinically but elicited a low level of CSP antibodies for a short duration. To provide a CSP-based candidate vaccine, we investigated recombinant CSP and NANP conjugates of various peptide lengths, with different N-terminal amino acids, bound at different ratios to various carrier proteins. Injected into mice, CSP alone and CSP or NANP conjugates induced antibodies with booster responses and were positive by the sporozoite immunofluorescent assay. The use of the mosquito stage P. falciparum ookinete surface protein, Pfs25, cross-linked onto itself as a carrier for NANP, induced in mice high levels of uniquely long-lasting antibodies to both vaccine components with secondary biological activities, that will provide immunity to liver infection by sporozoites and block transmission by mosquitoes.


Subject(s)
Malaria Vaccines/immunology , Peptides/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Animals , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Fluorescent Antibody Technique , Gas Chromatography-Mass Spectrometry , Malaria Vaccines/chemistry , Mice , Peptides/immunology , Protozoan Proteins/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , T-Lymphocytes/immunology
20.
Hum Vaccin ; 6(1): 90-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19806009

ABSTRACT

The RTS,S/AS01(E) malaria vaccine candidate has recently entered Phase 3 testing. Reaching this important milestone is the culmination of more than 20 years of research and development by GlaxoSmithKline and partners and collaborators. The vaccine has been developed to protect young children and infants living in Sub-Saharan Africa against clinical and severe disease caused by Plasmodium falciparum infection. Over the past 9 years, RTS,S/AS has been evaluated in multiple Phase 2 studies. The vaccine was shown to have a favorable safety profile and to be well tolerated in all age groups in which it was tested, including the intended target population of infants and young children in Sub-Saharan Africa. Data obtained so far suggest that RTS,S/AS can be co-administered with other vaccines included in the routine Expanded Program of Immunization (EPI). In Phase 2 testing, the vaccine candidate was shown to confer significant protection against P. falciparum infection and clinical disease, including severe malaria. Furthermore, a trend towards an indirect beneficial effect of the vaccine on non-malarial morbidities has been observed in several trials. In this paper, we will describe the genesis of the RTS,S/AS concept, including the rationale for selecting the circumsporozoite protein (CSP) as the target antigen. Early development history of the vaccine will be briefly described. We will present the most salient results from recent Phase 2 studies conducted in the target pediatric population, which have led to the decision to progress RTS,S/AS to Phase 3 testing. If the Phase 3 results confirm the observations made during Phase 2 testing, the RTS,S/AS vaccine, when broadly implemented and judiciously integrated with other malaria-prevention measures, would have a major public-health impact in Sub-Saharan Africa.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Africa South of the Sahara/epidemiology , Child, Preschool , Clinical Trials as Topic , Humans , Infant , Malaria Vaccines/adverse effects , Protozoan Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...