Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Tuberculosis (Edinb) ; 104: 46-57, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28454649

RESUMEN

M.bovis BCG vaccination against tuberculosis (TB) notoriously displays variable protective efficacy in different human populations. In non-human primate studies using rhesus macaques, despite efforts to standardise the model, we have also observed variable efficacy of BCG upon subsequent experimental M. tuberculosis challenge. In the present head-to-head study, we establish that the protective efficacy of standard parenteral BCG immunisation varies among different rhesus cohorts. This provides different dynamic ranges for evaluation of investigational vaccines, opportunities for identifying possible correlates of protective immunity and for determining why parenteral BCG immunisation sometimes fails. We also show that pulmonary mucosal BCG vaccination confers reduced local pathology and improves haematological and immunological parameters post-infection in animals that are not responsive to induction of protection by standard intra-dermal BCG. These results have important implications for pulmonary TB vaccination strategies in the future.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunogenicidad Vacunal , Mycobacterium tuberculosis/inmunología , Tuberculosis/prevención & control , Vacunación , Administración por Inhalación , Animales , Vacuna BCG/toxicidad , Modelos Animales de Enfermedad , Femenino , Inmunidad Mucosa , Inyecciones Intradérmicas , Macaca mulatta , Masculino , Mycobacterium tuberculosis/patogenicidad , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Factores de Tiempo , Tuberculosis/inmunología , Tuberculosis/microbiología
2.
PLoS One ; 11(10): e0164053, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27695087

RESUMEN

Plasmodium falciparum apical membrane antigen 1 (PfAMA1) is a leading asexual blood stage vaccine candidate for malaria. In preparation for clinical trials, three Diversity Covering (DiCo) PfAMA1 ectodomain proteins, designed to overcome the intrinsic polymorphism that is present in PfAMA1, were produced under Good Manufacturing Practice (GMP) in Pichia pastoris. Using identical methodology, the 3 strains were cultivated in 70-L scale fed-batch fermentations and PfAMA1-DiCos were purified by two chromatography steps, an ultrafiltration/diafiltration procedure and size exclusion chromatography, resulting in highly pure (>95%) PfAMA1-DiCo1, PfAMA1 DiCo2 and PfAMA1 DiCo3, with final yields of 1.8, 1.9 and 1.3 gram, respectively. N-terminal determinations showed that approximately 50% of each of the proteins lost 12 residues from their N-terminus, in accordance with SDS-PAGE (2 main bands) and MS-data. Under reducing conditions a site of limited proteolytic cleavage within a disulphide bonded region became evident. The three proteins quantitatively bound to the mAb 4G2 that recognizes a conformational epitope, suggesting proper folding of the proteins. The lyophilized Drug Product (1:1:1 mixture of PfAMA1-DiCo1, DiCo2, DiCo3) fulfilled all pre-set release criteria (appearance, dissolution rate, identity, purity, protein content, moisture content, sub-visible particles, immuno-potency (after reconstitution with adjuvant), abnormal toxicity, sterility and endotoxin), was stable in accelerated and real-time stability studies at -20°C for over 24 months. When formulated with adjuvants selected for clinical phase I evaluation, the Drug Product did not show adverse effect in a repeated-dose toxicity study in rabbits. The Drug Product has entered a phase Ia/Ib clinical trial.


Asunto(s)
Variación Antigénica , Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/biosíntesis , Vacunas contra la Malaria/inmunología , Proteínas de la Membrana/inmunología , Proteínas Protozoarias/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/biosíntesis , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Femenino , Fermentación , Humanos , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/efectos adversos , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Estabilidad Proteica , Proteínas Protozoarias/biosíntesis , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Control de Calidad , Conejos , Proteínas Recombinantes
3.
PLoS One ; 10(4): e0123567, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25886591

RESUMEN

The malaria parasite Plasmodium knowlesi, previously associated only with infection of macaques, is now known to infect humans as well and has become a significant public health problem in Southeast Asia. This species should therefore be targeted in vaccine and therapeutic strategies against human malaria. Apical Membrane Antigen 1 (AMA1), which plays a role in Plasmodium merozoite invasion of the erythrocyte, is currently being pursued in human vaccine trials against P. falciparum. Recent vaccine trials in macaques using the P. knowlesi orthologue PkAMA1 have shown that it protects against infection by this parasite species and thus should be developed for human vaccination as well. Here, we present the crystal structure of Domains 1 and 2 of the PkAMA1 ectodomain, and of its complex with the invasion-inhibitory monoclonal antibody R31C2. The Domain 2 (D2) loop, which is displaced upon binding the Rhoptry Neck Protein 2 (RON2) receptor, makes significant contacts with the antibody. R31C2 inhibits binding of the Rhoptry Neck Protein 2 (RON2) receptor by steric blocking of the hydrophobic groove and by preventing the displacement of the D2 loop which is essential for exposing the complete binding site on AMA1. R31C2 recognizes a non-polymorphic epitope and should thus be cross-strain reactive. PkAMA1 is much less polymorphic than the P. falciparum and P. vivax orthologues. Unlike these two latter species, there are no polymorphic sites close to the RON2-binding site of PkAMA1, suggesting that P. knowlesi has not developed a mechanism of immune escape from the host's humoral response to AMA1.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Protozoos/química , Proteínas de la Membrana/química , Plasmodium knowlesi/inmunología , Proteínas Protozoarias/química , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/inmunología , Secuencia de Bases , Cristalografía por Rayos X , Proteínas de la Membrana/inmunología , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Proteínas Protozoarias/inmunología
4.
Nat Commun ; 5: 4754, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25203297

RESUMEN

Plasmodium falciparum causes most human malaria deaths, having prehistorically evolved from parasites of African Great Apes. Here we explore the genomic basis of P. falciparum adaptation to human hosts by fully sequencing the genome of the closely related chimpanzee parasite species P. reichenowi, and obtaining partial sequence data from a more distantly related chimpanzee parasite (P. gaboni). The close relationship between P. reichenowi and P. falciparum is emphasized by almost complete conservation of genomic synteny, but against this strikingly conserved background we observe major differences at loci involved in erythrocyte invasion. The organization of most virulence-associated multigene families, including the hypervariable var genes, is broadly conserved, but P. falciparum has a smaller subset of rif and stevor genes whose products are expressed on the infected erythrocyte surface. Genome-wide analysis identifies other loci under recent positive selection, but a limited number of changes at the host-parasite interface may have mediated host switching.


Asunto(s)
Genoma/genética , Especificidad del Huésped/genética , Pan troglodytes/parasitología , Plasmodium falciparum/genética , Animales , Secuencia de Bases , Humanos/parasitología , Datos de Secuencia Molecular , Familia de Multigenes , Plasmodium/genética , Análisis de Secuencia de ADN
5.
PLoS One ; 9(1): e83704, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24421900

RESUMEN

The Block 2 region of the merozoite surface protein-1 (MSP-1) of Plasmodium falciparum has been identified as a target of protective immunity by a combination of seroepidemiology and parasite population genetics. Immunogenicity studies in small animals and Aotus monkeys were used to determine the efficacy of recombinant antigens derived from this region of MSP-1 as a potential vaccine antigen. Aotus lemurinus griseimembra monkeys were immunized three times with a recombinant antigen derived from the Block 2 region of MSP-1 of the monkey-adapted challenge strain, FVO of Plasmodium falciparum, using an adjuvant suitable for use in humans. Immunofluorescent antibody assays (IFA) against erythrocytes infected with P. falciparum using sera from the immunized monkeys showed that the MSP-1 Block 2 antigen induced significant antibody responses to whole malaria parasites. MSP-1 Block 2 antigen-specific enzyme-linked immunosorbent assays (ELISA) showed no significant differences in antibody titers between immunized animals. Immunized animals were challenged with the virulent P. falciparum FVO isolate and monitored for 21 days. Two out of four immunized animals were able to control their parasitaemia during the follow-up period, whereas two out of two controls developed fulminating parasitemia. Parasite-specific serum antibody titers measured by IFA were four-fold higher in protected animals than in unprotected animals. In addition, peptide-based epitope mapping of serum antibodies from immunized Aotus showed distinct differences in epitope specificities between protected and unprotected animals.


Asunto(s)
Formación de Anticuerpos/inmunología , Haplorrinos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/inmunología , Adyuvantes Inmunológicos , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos/inmunología , Antígenos de Protozoos/inmunología , Mapeo Epitopo , Epítopos/química , Epítopos/inmunología , Haplorrinos/sangre , Haplorrinos/parasitología , Humanos , Inmunización , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Datos de Secuencia Molecular , Parasitemia/inmunología , Parasitemia/parasitología , Proteínas Recombinantes/inmunología
6.
Antimicrob Agents Chemother ; 58(3): 1586-95, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24366744

RESUMEN

Preventing relapses of Plasmodium vivax malaria through a radical cure depends on use of the 8-aminoquinoline primaquine, which is associated with safety and compliance issues. For future malaria eradication strategies, new, safer radical curative compounds that efficiently kill dormant liver stages (hypnozoites) will be essential. A new compound with potential radical cure activity was identified using a low-throughput assay of in vitro-cultured hypnozoite forms of Plasmodium cynomolgi (an excellent and accessible model for Plasmodium vivax). In this assay, primary rhesus hepatocytes are infected with P. cynomolgi sporozoites, and exoerythrocytic development is monitored in the presence of compounds. Liver stage cultures are fixed after 6 days and stained with anti-Hsp70 antibodies, and the relative proportions of small (hypnozoite) and large (schizont) forms relative to the untreated controls are determined. This assay was used to screen a series of 18 known antimalarials and 14 new non-8-aminoquinolines (preselected for blood and/or liver stage activity) in three-point 10-fold dilutions (0.1, 1, and 10 µM final concentrations). A novel compound, designated KAI407 showed an activity profile similar to that of primaquine (PQ), efficiently killing the earliest stages of the parasites that become either primary hepatic schizonts or hypnozoites (50% inhibitory concentration [IC50] for hypnozoites, KAI407, 0.69 µM, and PQ, 0.84 µM; for developing liver stages, KAI407, 0.64 µM, and PQ, 0.37 µM). When given as causal prophylaxis, a single oral dose of 100 mg/kg of body weight prevented blood stage parasitemia in mice. From these results, we conclude that KAI407 may represent a new compound class for P. vivax malaria prophylaxis and potentially a radical cure.


Asunto(s)
Antimaláricos/farmacología , Imidazoles/farmacología , Malaria/tratamiento farmacológico , Plasmodium cynomolgi/efectos de los fármacos , Pirazinas/farmacología , Animales , Antimaláricos/uso terapéutico , Evaluación Preclínica de Medicamentos/métodos , Femenino , Hepatocitos/parasitología , Imidazoles/uso terapéutico , Técnicas In Vitro , Hígado/parasitología , Macaca mulatta/parasitología , Malaria/parasitología , Malaria/prevención & control , Ratones , Ratones Endogámicos ICR , Pirazinas/uso terapéutico , Esporozoítos/efectos de los fármacos
7.
Infect Immun ; 81(5): 1479-90, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23429538

RESUMEN

To overcome polymorphism in the malaria vaccine candidate Plasmodium falciparum apical membrane antigen 1 (PfAMA1), fusion protein chimeras comprised of three diversity-covering (DiCo) PfAMA1 molecules (D1, D2, and D3) and two allelic variants of the C-terminal 19-kDa region of merozoite surface protein 1 (MSP119) (variants M1 and M2) were generated. A mixture of fusion proteins (D1M1/D2M2D3) and the D1M1D2M2D3 fusion were compared to a single-unit mixture (D1/D2/D3/M1) in an immunological study in groups of rabbits. Following immunization, titers of antibodies (Abs) against four naturally occurring PfAMA1 alleles were high for all groups, as were growth inhibition assay (GIA) levels against two antigenically distinct laboratory parasite strains. Fusion of AMA1 to MSP119 did not suppress levels of antibodies against the AMA1 component. In addition, the breadth of antibody responses was unaffected. Anti-AMA1 antibodies were largely responsible for parasite growth inhibition, as shown in reversal-of-inhibition experiments by adding competing AMA1 antigen. For all groups, titration of the MSP119 antigen into the GIA led to only a small decrease in parasite inhibition, although titers of antibodies against MSP119 were increased 15-fold for the groups immunized with fusion proteins. GIA with affinity-purified anti-MSP119 antibodies showed that the 50% inhibitory concentrations of the anti-MSP119 antibody preparations were in the same order of magnitude for all animals tested, leading to the conclusion that fusing MSP119 to PfAMA1 leads to a small but significant increase in functional antibody levels. This study shows that combination of multiple vaccine candidates in fusion proteins may lead to improved characteristics of the vaccine.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Proteínas de la Membrana/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Modelos Animales de Enfermedad , Plasmodium falciparum/crecimiento & desarrollo , Conejos
8.
PLoS One ; 8(1): e54888, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23359816

RESUMEN

A major challenge for strategies to combat the human malaria parasite Plasmodium vivax is the presence of hypnozoites in the liver. These dormant forms can cause renewed clinical disease after reactivation through unknown mechanisms. The closely related non-human primate malaria P. cynomolgi is a frequently used model for studying hypnozoite-induced relapses. Here we report the generation of the first transgenic P. cynomolgi parasites that stably express fluorescent markers in liver stages by transfection with novel DNA-constructs containing a P. cynomolgi centromere. Analysis of fluorescent liver stages in culture identified, in addition to developing liver-schizonts, uninucleate persisting parasites that were atovaquone resistant but primaquine sensitive, features associated with hypnozoites. We demonstrate that these hypnozoite-forms could be isolated by fluorescence-activated cell sorting. The fluorescently-tagged parasites in combination with FACS-purification open new avenues for a wide range of studies for analysing hypnozoite biology and reactivation.


Asunto(s)
Antimaláricos/farmacología , Hígado/parasitología , Plasmodium cynomolgi/fisiología , Animales , Animales Modificados Genéticamente , Atovacuona/farmacología , Fluorescencia , Humanos , Plasmodium cynomolgi/efectos de los fármacos , Plasmodium cynomolgi/crecimiento & desarrollo , Primaquina/farmacología
9.
Mol Cell Proteomics ; 12(2): 426-48, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23197789

RESUMEN

Malaria parasites actively remodel the infected red blood cell (irbc) by exporting proteins into the host cell cytoplasm. The human parasite Plasmodium falciparum exports particularly large numbers of proteins, including proteins that establish a vesicular network allowing the trafficking of proteins onto the surface of irbcs that are responsible for tissue sequestration. Like P. falciparum, the rodent parasite P. berghei ANKA sequesters via irbc interactions with the host receptor CD36. We have applied proteomic, genomic, and reverse-genetic approaches to identify P. berghei proteins potentially involved in the transport of proteins to the irbc surface. A comparative proteomics analysis of P. berghei non-sequestering and sequestering parasites was used to determine changes in the irbc membrane associated with sequestration. Subsequent tagging experiments identified 13 proteins (Plasmodium export element (PEXEL)-positive as well as PEXEL-negative) that are exported into the irbc cytoplasm and have distinct localization patterns: a dispersed and/or patchy distribution, a punctate vesicle-like pattern in the cytoplasm, or a distinct location at the irbc membrane. Members of the PEXEL-negative BIR and PEXEL-positive Pb-fam-3 show a dispersed localization in the irbc cytoplasm, but not at the irbc surface. Two of the identified exported proteins are transported to the irbc membrane and were named erythrocyte membrane associated proteins. EMAP1 is a member of the PEXEL-negative Pb-fam-1 family, and EMAP2 is a PEXEL-positive protein encoded by a single copy gene; neither protein plays a direct role in sequestration. Our observations clearly indicate that P. berghei traffics a diverse range of proteins to different cellular locations via mechanisms that are analogous to those employed by P. falciparum. This information can be exploited to generate transgenic humanized rodent P. berghei parasites expressing chimeric P. berghei/P. falciparum proteins on the surface of rodent irbc, thereby opening new avenues for in vivo screening adjunct therapies that block sequestration.


Asunto(s)
Malaria/metabolismo , Plasmodium berghei/genética , Proteoma/genética , Proteínas Protozoarias/genética , Esquizontes/metabolismo , Trofozoítos/metabolismo , Animales , Antígenos CD36/química , Antígenos CD36/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes , Interacciones Huésped-Parásitos , Luciferasas , Malaria/parasitología , Ratones , Mutación , Plasmodium berghei/química , Plasmodium berghei/metabolismo , Transporte de Proteínas , Proteoma/química , Proteoma/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Esquizontes/química , Espectrometría de Masas en Tándem , Transfección , Trofozoítos/química
10.
PLoS One ; 7(6): e38898, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768052

RESUMEN

UNLABELLED: Plasmodium falciparum: apical membrane antigen 1 (AMA1) is a candidate malaria vaccine antigen expressed on merozoites and sporozoites. The polymorphic nature of AMA1 may compromise vaccine induced protection. The humoral response induced by two dosages (10 and 50 µg) of a single allele AMA1 antigen (FVO) formulated with Alhydrogel, Montanide ISA 720 or AS02 was investigated in 47 malaria-naïve adult volunteers. Volunteers were vaccinated 3 times at 4 weekly intervals and serum samples obtained four weeks after the third immunization were analysed for (i) Antibody responses to various allelic variants, (ii) Domain specificity, (iii) Avidity, (iv) IgG subclass levels, by ELISA and (v) functionality of antibody responses by Growth Inhibition Assay (GIA). About half of the antibodies induced by vaccination cross reacted with heterologous AMA1 alleles. The choice of adjuvant determined the magnitude of the antibody response, but had only a marginal influence on specificity, avidity, domain recognition or subclass responses. The highest antibody responses were observed for AMA1 formulated with AS02. The Growth Inhibition Assay activity of the antibodies was proportional to the amount of antigen specific IgG and the functional capacity of the antibodies was similar for heterologous AMA1-expressing laboratory strains. TRIAL REGISTRATION: ClinicalTrials.gov NCT00730782.


Asunto(s)
Alelos , Antígenos de Protozoos/inmunología , Salud , Inmunidad Humoral/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Adulto , Secuencia de Aminoácidos , Afinidad de Anticuerpos/inmunología , Antígenos de Protozoos/sangre , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina G/clasificación , Inmunoglobulina G/inmunología , Malaria Falciparum/sangre , Malaria Falciparum/prevención & control , Masculino , Proteínas de la Membrana/sangre , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Estructura Terciaria de Proteína , Proteínas Protozoarias/sangre , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Alineación de Secuencia , Volumetría , Vacunación , Adulto Joven
11.
J Exp Med ; 209(1): 93-107, 2012 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-22184632

RESUMEN

Adherence of parasite-infected red blood cells (irbc) to the vascular endothelium of organs plays a key role in the pathogenesis of Plasmodium falciparum malaria. The prevailing hypothesis of why irbc adhere and sequester in tissues is that this acts as a mechanism of avoiding spleen-mediated clearance. Irbc of the rodent parasite Plasmodium berghei ANKA sequester in a fashion analogous to P. falciparum by adhering to the host receptor CD36. To experimentally determine the significance of sequestration for parasite growth, we generated a mutant P. berghei ANKA parasite with a reduced CD36-mediated adherence. Although the cognate parasite ligand binding to CD36 is unknown, we show that nonsequestering parasites have reduced growth and we provide evidence that in addition to avoiding spleen removal, other factors related to CD36-mediated sequestration are beneficial for parasite growth. These results reveal for the first time the importance of sequestration to a malaria infection, with implications for the development of strategies aimed at reducing pathology by inhibiting tissue sequestration.


Asunto(s)
Antígenos CD36/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Malaria/metabolismo , Malaria/parasitología , Plasmodium berghei/metabolismo , Animales , Antígenos CD36/genética , Adhesión Celular/genética , Ciclo Celular/genética , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo , Transporte de Proteínas , Proteómica , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Ratas , Ratas Wistar , Esquizontes/metabolismo , Esplenectomía
12.
Infect Immun ; 80(3): 1280-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22202121

RESUMEN

Merozoite surface protein 1 (MSP1) is a target for malaria vaccine development. Antibodies to the 19-kDa carboxy-terminal region referred to as MSP1(19) inhibit erythrocyte invasion and parasite growth, with some MSP1-specific antibodies shown to inhibit the proteolytic processing of MSP1 that occurs at invasion. We investigated a series of antibodies purified from rabbits immunized with MSP1(19) and AMA1 recombinant proteins for their ability to inhibit parasite growth, initially looking at MSP1 processing. Although significant inhibition of processing was mediated by several of the antibody samples, there was no clear relationship with overall growth inhibition by the same antibodies. However, no antibody samples inhibited processing but not invasion, suggesting that inhibition of MSP1 processing contributes to but is not the only mechanism of antibody-mediated inhibition of invasion and growth. Examining other mechanisms by which MSP1-specific antibodies inhibit parasite growth, we show that MSP1(19)-specific antibodies are taken up into invaded erythrocytes, where they persist for significant periods and result in delayed intracellular parasite development. This delay may result from antibody interference with coalescence of MSP1(19)-containing vesicles with the food vacuole. Antibodies raised against a modified recombinant MSP1(19) sequence were more efficient at delaying intracellular growth than those to the wild-type protein. We propose that antibodies specific for MSP1(19) can mediate inhibition of parasite growth by at least three mechanisms: inhibition of MSP1 processing, direct inhibition of invasion, and inhibition of parasite development following invasion. The balance between mechanisms may be modulated by modifying the immunogen used to induce the antibodies.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Proteína 1 de Superficie de Merozoito/metabolismo , Merozoítos/crecimiento & desarrollo , Merozoítos/inmunología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Animales , Eritrocitos/parasitología , Conejos
13.
PLoS One ; 6(10): e26616, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22073118

RESUMEN

Polymorphic parasite antigens are known targets of protective immunity to malaria, but this antigenic variation poses challenges to vaccine development. A synthetic MSP-1 Block 2 construct, based on all polymorphic variants found in natural Plasmodium falciparum isolates has been designed, combined with the relatively conserved Block 1 sequence of MSP-1 and expressed in E.coli. The MSP-1 Hybrid antigen has been produced with high yield by fed-batch fermentation and purified without the aid of affinity tags resulting in a pure and extremely thermostable antigen preparation. MSP-1 hybrid is immunogenic in experimental animals using adjuvants suitable for human use, eliciting antibodies against epitopes from all three Block 2 serotypes. Human serum antibodies from Africans naturally exposed to malaria reacted to the MSP-1 hybrid as strongly as, or better than the same serum reactivities to individual MSP-1 Block 2 antigens, and these antibody responses showed clear associations with reduced incidence of malaria episodes. The MSP-1 hybrid is designed to induce a protective antibody response to the highly polymorphic Block 2 region of MSP-1, enhancing the repertoire of MSP-1 Block 2 antibody responses found among immune and semi-immune individuals in malaria endemic areas. The target population for such a vaccine is young children and vulnerable adults, to accelerate the acquisition of a full range of malaria protective antibodies against this polymorphic parasite antigen.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Inmunoglobulina G/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/inmunología , Adolescente , Adulto , Animales , Anticuerpos Monoclonales/inmunología , Estudios de Casos y Controles , Niño , Preescolar , Estudios Transversales , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunización , Immunoblotting , Macaca mulatta , Malaria Falciparum/prevención & control , Ratones , Ratones Endogámicos DBA , Plasmodium falciparum/crecimiento & desarrollo , Conejos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
14.
Malar J ; 10: 182, 2011 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-21726452

RESUMEN

BACKGROUND: Increasing the breadth of the functional antibody response through immunization with Plasmodium falciparum apical membrane antigen 1 (PfAMA1) multi-allele vaccine formulations has been demonstrated in several rodent and rabbit studies. This study assesses the safety and immunogenicity of three PfAMA1 Diversity-Covering (DiCo) vaccine candidates formulated as an equimolar mixture (DiCo mix) in CoVaccine HT™ or Montanide ISA 51, as well as that of a PfAMA1-MSP119 fusion protein formulated in Montanide ISA 51. METHODS: Vaccine safety in rhesus macaques was monitored by animal behaviour observation and assessment of organ and systemic functions through clinical chemistry and haematology measurements. The immunogenicity of vaccine formulations was assessed by enzyme-linked immunosorbent assays and in vitro parasite growth inhibition assays with three culture-adapted P. falciparum strains. RESULTS: These data show that both adjuvants were well tolerated with only transient changes in a few of the chemical and haematological parameters measured. DiCo mix formulated in CoVaccine HT™ proved immunologically and functionally superior to the same candidate formulated in Montanide ISA 51. Immunological data from the fusion protein candidate was however difficult to interpret as four out of six immunized animals were non-responsive for unknown reasons. CONCLUSIONS: The study highlights the safety and immunological benefits of DiCo mix as a potential human vaccine against blood stage malaria, especially when formulated in CoVaccine HT™, and adds to the accumulating data on the specificity broadening effects of DiCo mix.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/efectos adversos , Vacunas contra la Malaria/inmunología , Manitol/análogos & derivados , Proteínas de la Membrana/inmunología , Ácidos Oléicos/administración & dosificación , Ácidos Oléicos/efectos adversos , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Supervivencia Celular , Femenino , Humanos , Inmunoglobulina G/sangre , Macaca mulatta , Vacunas contra la Malaria/administración & dosificación , Masculino , Manitol/administración & dosificación , Manitol/efectos adversos , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Proteínas Recombinantes de Fusión/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/inmunología
15.
PLoS One ; 6(5): e20547, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21655233

RESUMEN

Plasmodium falciparum apical membrane antigen 1 (PfAMA1) is a leading blood stage vaccine candidate. Plasmodium knowlesi AMA1 (PkAMA1) was produced and purified using similar methodology as for clinical grade PfAMA1 yielding a pure, conformational intact protein. Combined with the adjuvant CoVaccine HT™, PkAMA1 was found to be highly immunogenic in rabbits and the efficacy of the PkAMA1 was subsequently tested in a rhesus macaque blood-stage challenge model. Six rhesus monkeys were vaccinated with PkAMA1 and a control group of 6 were vaccinated with PfAMA1. A total of 50 µg AMA1 was administered intramuscularly three times at 4 week intervals. One of six rhesus monkeys vaccinated with PkAMA1 was able to control parasitaemia, upon blood stage challenge with P. knowlesi H-strain. Four out of the remaining five showed a delay in parasite onset that correlated with functional antibody titres. In the PfAMA1 vaccinated control group, five out of six animals had to be treated with antimalarials 8 days after challenge; one animal did not become patent during the challenge period. Following a rest period, animals were boosted and challenged again. Four of the six rhesus monkeys vaccinated with PkAMA1 were able to control the parasitaemia, one had a delayed onset of parasitaemia and one animal was not protected, while all control animals required treatment. To confirm that the control of parasitaemia was AMA1-related, animals were allowed to recover, boosted and re-challenged with P. knowlesi Nuri strain. All control animals had to be treated with antimalarials by day 8, while five out of six PkAMA1 vaccinated animals were able to control parasitaemia. This study shows that: i) Yeast-expressed PkAMA1 can protect against blood stage challenge; ii) Functional antibody levels as measured by GIA correlated inversely with the day of onset and iii) GIA IC(50) values correlated with estimated in vivo growth rates.


Asunto(s)
Macaca mulatta/inmunología , Macaca mulatta/parasitología , Vacunas contra la Malaria/inmunología , Plasmodium knowlesi/inmunología , Vacunación/métodos , Adyuvantes Inmunológicos , Animales , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática
16.
PLoS One ; 6(3): e18162, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21483865

RESUMEN

BACKGROUND: Amongst the Plasmodium species in humans, only P. vivax and P. ovale produce latent hepatic stages called hypnozoites, which are responsible for malaria episodes long after a mosquito bite. Relapses contribute to increased morbidity, and complicate malaria elimination programs. A single drug effective against hypnozoites, primaquine, is available, but its deployment is curtailed by its haemolytic potential in glucose-6-phosphate dehydrogenase deficient persons. Novel compounds are thus urgently needed to replace primaquine. Discovery of compounds active against hypnozoites is restricted to the in vivo P. cynomolgi-rhesus monkey model. Slow growing hepatic parasites reminiscent of hypnozoites had been noted in cultured P. vivax-infected hepatoma cells, but similar forms are also observed in vitro by other species including P. falciparum that do not produce hypnozoites. METHODOLOGY: P. falciparum or P. cynomolgi sporozoites were used to infect human or Macaca fascicularis primary hepatocytes, respectively. The susceptibility of the slow and normally growing hepatic forms obtained in vitro to three antimalarial drugs, one active against hepatic forms including hypnozoites and two only against the growing forms, was measured. RESULTS: The non-dividing slow growing P. cynomolgi hepatic forms, observed in vitro in primary hepatocytes from the natural host Macaca fascicularis, can be distinguished from similar forms seen in P. falciparum-infected human primary hepatocytes by the differential action of selected anti-malarial drugs. Whereas atovaquone and pyrimethamine are active on all the dividing hepatic forms observed, the P. cynomolgi slow growing forms are highly resistant to treatment by these drugs, but remain susceptible to primaquine. CONCLUSION: Resistance of the non-dividing P. cynomolgi forms to atovaquone and pyrimethamine, which do not prevent relapses, strongly suggests that these slow growing forms are hypnozoites. This represents a first step towards the development of a practical medium-throughput in vitro screening assay for novel hypnozoiticidal drugs.


Asunto(s)
Hepatocitos/parasitología , Plasmodium/fisiología , Esporozoítos/fisiología , Animales , Antimaláricos/farmacología , Células Cultivadas , Humanos , Macaca fascicularis , Plasmodium/efectos de los fármacos , Esporozoítos/efectos de los fármacos
17.
Malar J ; 10: 40, 2011 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-21320299

RESUMEN

BACKGROUND: Antibodies to key Plasmodium falciparum surface antigens have been shown to be important effectors that mediate clinical immunity to malaria. The cross-strain fraction of anti-malarial antibodies may however be required to achieve strain-transcending immunity. Such antibody responses against Plasmodium falciparum apical membrane antigen 1 (PfAMA1), a vaccine target molecule that is expressed in both liver and blood stages of the parasite, can be elicited through immunization with a mixture of allelic variants of the parasite molecule. Cross-strain antibodies are most likely elicited against epitopes that are shared by the allelic antigens in the vaccine cocktail. METHODS: A standard competition ELISA was used to address whether the antibody response can be further focused on shared epitopes by exclusively boosting these common determinants through immunization of rabbits with different PfAMA1 alleles in sequence. The in vitro parasite growth inhibition assay was used to further evaluate the functional effects of the broadened antibody response that is characteristic of multi-allele vaccine strategies. RESULTS: A mixed antigen immunization protocol elicited humoral responses that were functionally similar to those elicited by a sequential immunization protocol (p > 0.05). Sequential exposure to the different PfAMA1 allelic variants induced immunological recall of responses to previous alleles and yielded functional cross-strain antibodies that would be capable of optimal growth inhibition of variant parasites at high enough concentrations. CONCLUSIONS: These findings may have implications for the current understanding of the natural acquisition of clinical immunity to malaria as well as for rational vaccine design.


Asunto(s)
Antígenos de Protozoos/inmunología , Inmunización/métodos , Vacunas contra la Malaria/inmunología , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/administración & dosificación , Protección Cruzada , Ensayo de Inmunoadsorción Enzimática/métodos , Epítopos/inmunología , Vacunas contra la Malaria/administración & dosificación , Proteínas de la Membrana/administración & dosificación , Proteínas Protozoarias/administración & dosificación , Conejos
18.
Malar J ; 10: 29, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21303498

RESUMEN

BACKGROUND: A DNA prime, poxvirus (COPAK) boost vaccination regime with four antigens, i.e. a combination of two Plasmodium knowlesi sporozoite (csp/ssp2) and two blood stage (ama1/msp142) genes, leads to self-limited parasitaemia in 60% of rhesus monkeys and survival from an otherwise lethal infection with P. knowlesi. In the present study, the role of the blood stage antigens in protection was studied in depth, focusing on antibody formation against the blood stage antigens and the functionality thereof. METHODS: Rhesus macaques were immunized with the four-component vaccine and subsequently challenged i.v. with 100 P. knowlesi sporozoites. During immunization and challenge, antibody titres against the two blood stage antigens were determined, as well as the in vitro growth inhibition capacity of those antibodies. Antigen reversal experiments were performed to determine the relative contribution of antibodies against each of the two blood stage antigens to the inhibition. RESULTS: After vaccination, PkAMA1 and PkMSP119 antibody titres in vaccinated animals were low, which was reflected in low levels of inhibition by these antibodies as determined by in vitro inhibition assays. Interestingly, after sporozoite challenge antibody titres against blood stage antigens were boosted over 30-fold in both protected and not protected animals. The in vitro inhibition levels increased to high levels (median inhibitions of 59% and 56% at 6 mg/mL total IgG, respectively). As growth inhibition levels were not significantly different between protected and not protected animals, the ability to control infection appeared cannot be explained by GIA levels. Judged by in vitro antigen reversal growth inhibition assays, over 85% of the inhibitory activity of these antibodies was directed against PkAMA1. CONCLUSIONS: This is the first report that demonstrates that a DNA prime/poxvirus boost vaccination regimen induces low levels of malaria parasite growth inhibitory antibodies, which are boosted to high levels upon challenge. No association could, however, be established between the levels of inhibitory capacity in vitro and protection, either after vaccination or after challenge.


Asunto(s)
Macaca mulatta/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Plasmodium knowlesi/genética , Poxviridae/genética , Animales , Anticuerpos Antiprotozoarios , Antígenos de Protozoos/sangre , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Ensayo de Inmunoadsorción Enzimática , Inmunización Secundaria , Macaca mulatta/sangre , Malaria/sangre , Malaria/prevención & control , Vacunas contra la Malaria/química , Plásmidos/metabolismo , Plasmodium knowlesi/inmunología , Proteínas Protozoarias/sangre , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Resultado del Tratamiento
19.
PLoS Pathog ; 7(2): e1001276, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21347343

RESUMEN

Obligate intracellular Apicomplexa parasites share a unique invasion mechanism involving a tight interaction between the host cell and the parasite surfaces called the moving junction (MJ). The MJ, which is the anchoring structure for the invasion process, is formed by secretion of a macromolecular complex (RON2/4/5/8), derived from secretory organelles called rhoptries, into the host cell membrane. AMA1, a protein secreted from micronemes and associated with the parasite surface during invasion, has been shown in vitro to bind the MJ complex through a direct association with RON2. Here we show that RON2 is inserted as an integral membrane protein in the host cell and, using several interaction assays with native or recombinant proteins, we define the region that binds AMA1. Our studies were performed both in Toxoplasma gondii and Plasmodium falciparum and although AMA1 and RON2 proteins have diverged between Apicomplexa species, we show an intra-species conservation of their interaction. More importantly, invasion inhibition assays using recombinant proteins demonstrate that the RON2-AMA1 interaction is crucial for both T. gondii and P. falciparum entry into their host cells. This work provides the first evidence that AMA1 uses the rhoptry neck protein RON2 as a receptor to promote invasion by Apicomplexa parasites.


Asunto(s)
Antígenos de Protozoos/metabolismo , Apicomplexa/fisiología , Interacciones Huésped-Parásitos/fisiología , Proteínas Protozoarias/metabolismo , Internalización del Virus , Animales , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Apicomplexa/genética , Apicomplexa/metabolismo , Células Cultivadas , Chlorocebus aethiops , Conexinas/metabolismo , Secuencia Conservada , Interacciones Huésped-Parásitos/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Modelos Biológicos , Modelos Moleculares , Parásitos/genética , Parásitos/metabolismo , Parásitos/fisiología , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiología , Unión Proteica/genética , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasma/fisiología , Células Vero
20.
PLoS One ; 5(11): e15391, 2010 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21082025

RESUMEN

There is increasing interest in multi-allele vaccines to overcome strain-specificity against polymorphic vaccine targets such as Apical Membrane Antigen 1 (AMA1). These have been shown to induce broad inhibitory antibodies in vitro and formed the basis for the design of three Diversity-Covering (DiCo) proteins with similar immunological effects. The antibodies produced are to epitopes that are shared between vaccine alleles and theoretically, increasing the number of component AMA1 alleles is expected to broaden the antibody response. A plateau effect could however impose a limit on the number of alleles needed to achieve the broadest specificity. Moreover, production cost and the vaccine formulation process would limit the number of component alleles. In this paper, we compare rabbit antibody responses elicited with multi-allele vaccines incorporating seven (three DiCos and four natural AMA1 alleles) and three (DiCo mix) antigens for gains in broadened specificity. We also investigate the effect of three adjuvant platforms on antigen specificity and antibody functionality. Our data confirms a broadened response after immunisation with DiCo mix in all three adjuvants. Higher antibody titres were elicited with either CoVaccine HT™ or Montanide ISA 51, resulting in similar in vitro inhibition (65-82%) of five out of six culture-adapted P. falciparum strains. The antigen binding specificities of elicited antibodies were also similar and independent of the adjuvant used or the number of vaccine component alleles. Thus neither the four extra antigens nor adjuvant had any observable benefits with respect to specificity broadening, although adjuvant choice influenced the absolute antibody levels and thus the extent of parasite inhibition. Our data confirms the feasibility and potential of multi-allele PfAMA1 formulations, and highlights the need for adjuvants with improved antibody potentiation properties for AMA1-based vaccines.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Alelos , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Ensayo de Inmunoadsorción Enzimática , Estudios de Factibilidad , Inmunidad Humoral/inmunología , Inmunización/métodos , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/prevención & control , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Conejos , Homología de Secuencia de Aminoácido , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA