Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Struct Mol Biol ; 13(1): 90-1, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16327807

RESUMO

P25 and P28 proteins are essential for Plasmodium parasites to infect mosquitoes and are leading candidates for a transmission-blocking malaria vaccine. The Plasmodium vivax P25 is a triangular prism that could tile the parasite surface. The residues forming the triangle are conserved in P25 and P28 from all Plasmodium species. A cocrystal structure shows that a transmission-blocking antibody uses only its heavy chain to bind Pvs25 at a vertex of the triangle.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Antígenos de Superfície/química , Antígenos de Superfície/metabolismo , Culicidae/parasitologia , Vacinas Antimaláricas/química , Vacinas Antimaláricas/metabolismo , Plasmodium vivax/química , Plasmodium vivax/crescimento & desenvolvimento , Animais , Modelos Moleculares , Plasmodium vivax/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
2.
Infect Immun ; 77(2): 817-24, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19015251

RESUMO

It is well known that exposure to one antigen can modulate the immune responses that develop following exposure to closely related antigens. It is also known that the composition of the repertoire can be skewed to favor epitopes shared between a current infection and a preceding one, a phenomenon referred to as "original antigenic sin." It was of interest, therefore, to investigate the antibody response that develops following exposure to the malaria vaccine candidate homologue Plasmodium yoelii MSP1(19) in mice that had previously experienced malaria infection and vice versa. In this study, preexposure of mice to Plasmodium yoelii elicited native anti-MSP1(19) antibody responses, which could be boosted by vaccination with recombinant MSP1(19). Likewise, infection of MSP1(19)-primed mice with P. yoelii led to an increase of anti-MSP1(19) antibodies. However, this increase was at the expense of antibodies to parasite determinants other than MSP1(19). This change in the balance of antibody specificities significantly affected the ability of mice to withstand a subsequent infection. These data have particular relevance to the possible outcome of malaria vaccination for those situations where the vaccine response is suboptimal and suggest that suboptimal vaccination may in fact render the ultimate acquisition of natural immunity more difficult.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium yoelii/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Feminino , Humanos , Vacinas Antimaláricas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
3.
Infect Immun ; 77(12): 5496-500, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19752035

RESUMO

Malaria vaccines based on ookinete surface proteins (OSPs) of the malaria parasites block oocyst development in feeding mosquitoes and hence disrupt the parasite life cycle and prevent the disease from being transmitted to other individuals. To investigate whether a noninvasive mucosal vaccination regimen effectively blocks parasite transmission in vivo, Plasmodium yoelii Pys25, a homolog of the Pfs25 and Pvs25 OSPs of Plasmodium falciparum and Plasmodium vivax, respectively, was intranasally (i.n.) administered using a complement-deficient DBA/2 mouse malaria infection model, in which a highly elevated level of oocysts develops in feeding mosquitoes. Vaccinated mice developed a robust antibody response when the vaccine antigen was given together with cholera toxin adjuvant. The induced immune serum was passively transferred to DBA/2 mice 3 days after infection with P. yoelii 17XL, and Anopheles stephensi mosquitoes were allowed to feed on the infected mice before or after serum transfusion. This passive immunization completely blocked oocyst development; however, immune serum induced by the antigen or adjuvant alone did not have such a profound antiparasite effect. Further, when i.n. vaccinated mice were infected with the parasite and then mosquitoes were allowed to directly feed on the infected mice, complete blockage of transmission was again observed. To our knowledge, this is the first time that mucosal vaccination has been demonstrated to be efficacious for directly preventing parasite transmission from vaccinated animals to mosquitoes, and the results may provide important insight into rational design of nonparenteral vaccines for use against human malaria.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Malária/transmissão , Proteínas de Protozoários/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anopheles/fisiologia , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/administração & dosagem , Toxina da Cólera/administração & dosagem , Vetores de Doenças , Feminino , Vacinas Antimaláricas/administração & dosagem , Camundongos , Camundongos Endogâmicos DBA , Plasmodium yoelii/imunologia , Proteínas de Protozoários/administração & dosagem
4.
Immunol Lett ; 81(1): 13-24, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11841841

RESUMO

We have constructed a DNA plasmid vaccine encoding the C-terminal 42-kDa region of the merozoite surface protein 1 (pMSP1(42)) from the 3D7 strain of Plasmodium falciparum (Pf3D7). This plasmid expressed recombinant MSP1(42) after in vitro transfection in mouse VM92 cells. Rhesus monkeys immunized with pMSP1(42) produced antibodies reactive with Pf3D7 infected erythrocytes by IFAT, and by ELISA against yeast produced MSP1(19) (yMSP1(19)). Immunization also induced antigen specific T cell responses as measured by interferon-gamma production, and by classical CTL chromium release assays. In addition, immunization with pMSP1(42) primed animals for an enhanced antibody response to a subsequent boost with the recombinant yMSP1(19). We also evaluated Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) as an adjuvant for pMSP1(42.) We tested both rhesus GM-CSF expressed from a DNA plasmid, and E. coli produced recombinant human GM-CSF. Plasmids encoding rhesus GM-CSF (prhGM-CSF) and human GM-CSF (phuGM-CSF) were constructed; these plasmids expressed bio-active recombinant GMCSF. Co-immunization with a mixture of prhGM-CSF and pMSP1(42) induced higher specific antibody responses after the first dose of plasmid, but after three doses of DNA monkeys immunized with or without prhGM-CSF had the same final antibody titers and T cell responses. In comparison, rhuGM-CSF protein did not lead to accelerated antibody production after the first DNA dose. However, antibody titers were maintained at a slightly higher level in monkeys receiving GM-CSF protein, and they had a higher response to boosting with recombinant MSP1(19). The GM-CSF plasmid or protein appears to be less potent as an adjuvant in rhesus monkeys than each is in mice, and more work is needed to determine if GM-CSF can be a useful adjuvant in DNA vaccination of primates.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Vacinas Antimaláricas , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Vacinas de DNA , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antiprotozoários/imunologia , Citotoxicidade Imunológica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interferon gama/imunologia , Macaca mulatta , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/imunologia , Plasmídeos , Plasmodium falciparum/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia
5.
Am J Trop Med Hyg ; 69(5): 536-41, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14695092

RESUMO

We have previously demonstrated that mouse antisera against yeast-produced recombinant forms of the ookinete surface proteins of Plasmodium vivax (Pvs25 and Pvs28) blocks transmission of the homologous P. vivax (Sal I strain). In this study, we developed mouse and rabbit antisera against Pvs25 and Pvs28 and evaluated the efficacy of these vaccine candidates against natural isolates of P. vivax in Thailand. Although both Pvs25 and Pvs28 genes are polymorphic, sera from mice immunized using alum adjuvant completely inhibited oocyst development for most human isolates, whereas sera from rabbits immunized with either alum or Freund's adjuvant were partially inhibitory. All inhibition occurred in an antibody dose dependent fashion. Data from this study clearly demonstrates that antibodies raised against Sal I-based vaccines overcome the genetic polymorphism of Pvs25 and Pvs28 present in natural isolates of P. vivax, suggesting the wide range applicability of Sal I based vaccines.


Assuntos
Anopheles/parasitologia , Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Adolescente , Adulto , Animais , Antígenos de Protozoários/imunologia , Antígenos de Superfície/imunologia , DNA de Protozoário/genética , Feminino , Humanos , Injeções Intraperitoneais , Insetos Vetores/parasitologia , Malária Vivax/transmissão , Camundongos , Camundongos Endogâmicos , Plasmodium vivax/genética , Coelhos , Proteínas Recombinantes/imunologia , Tailândia
6.
PLoS Clin Trials ; 2(4): e12, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17415408

RESUMO

OBJECTIVES: To assess the safety and immunogenicity of two vaccines, MSP1(42)-FVO/Alhydrogel and MSP1(42)-3D7/Alhydrogel, targeting blood-stage Plasmodium falciparum parasites. DESIGN: A Phase 1 open-label, dose-escalating study. SETTING: Quintiles Phase 1 Services, Lenexa, Kansas between July 2004 and November 2005. PARTICIPANTS: Sixty healthy malaria-naïve volunteers 18-48 y of age. INTERVENTIONS: The C-terminal 42-kDa region of merozoite surface protein 1 (MSP1(42)) corresponding to the two allelic forms present in FVO and 3D7 P. falciparum lines were expressed in Escherichia coli, refolded, purified, and formulated on Alhydrogel (aluminum hydroxide). For each vaccine, volunteers in each of three dose cohorts (5, 20, and 80 microg) were vaccinated at 0, 28, and 180 d. Volunteers were followed for 1 y. OUTCOME MEASURES: The safety of MSP1(42)-FVO/Alhydrogel and MSP1(42)-3D7/Alhydrogel was assessed. The antibody response to each vaccine was measured by reactivity to homologous and heterologous MSP1(42), MSP1(19), and MSP1(33) recombinant proteins and recognition of FVO and 3D7 parasites. RESULTS: Anti-MSP1(42) antibodies were detected by ELISA in 20/27 (74%) and 22/27 (81%) volunteers receiving three vaccinations of MSP1(42)-FVO/Alhydrogel or MSP1(42)-3D7/Alhydrogel, respectively. Regardless of the vaccine, the antibodies were cross-reactive to both MSP1(42)-FVO and MSP1(42)-3D7 proteins. The majority of the antibody response targeted the C-terminal 19-kDa domain of MSP1(42), although low-level antibodies to the N-terminal 33-kDa domain of MSP1(42) were also detected. Immunofluorescence microscopy of sera from the volunteers demonstrated reactivity with both FVO and 3D7 P. falciparum schizonts and free merozoites. Minimal in vitro growth inhibition of FVO or 3D7 parasites by purified IgG from the sera of the vaccinees was observed. CONCLUSIONS: The MSP1(42)/Alhydrogel vaccines were safe and well tolerated but not sufficiently immunogenic to generate a biologic effect in vitro. Addition of immunostimulants to the Alhydrogel formulation to elicit higher vaccine-induced responses in humans may be required for an effective vaccine.

7.
Infect Immun ; 73(7): 3963-70, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15972483

RESUMO

Recombinant apical membrane antigen 1 (AMA1) is a leading vaccine candidate for Plasmodium falciparum malaria, as antibodies against recombinant P. falciparum AMA1 (PfAMA1) interrupt merozoite invasion into erythrocytes. In order to investigate the role of posttranslational modification in modulating the functional immune response to recombinant AMA1, two separate alleles of PfAMA1 (FVO and 3D7), in which native N-glycosylation sites have been mutated, were produced using Escherichia coli and a Pichia pastoris expression system. Recombinant Pichia pastoris AMA1-FVO (PpAMA1-FVO) and PpAMA1-3D7 are O-linked glycosylated, and 45% of PpAMA1-3D7 is nicked, though all four recombinant molecules react with conformation-specific monoclonal antibodies. To address the immunological effect of O-linked glycosylation, we compared the immunogenicity of E. coli AMA1-FVO (EcAMA1-FVO) and PpAMA1-FVO antigens, since both molecules are intact. The effect of antigen nicking was then investigated by comparing the immunogenicity of EcAMA1-3D7 and PpAMA1-3D7. Our data demonstrate that there is no significant difference in the rabbit antibody titer elicited towards EcAMA1-FVO and PpAMA1-FVO or to EcAMA1-3D7 and PpAMA1-3D7. Furthermore, we have demonstrated that recombinant AMA1 (FVO or 3D7), whether expressed and refolded from E. coli or produced from the Pichia expression system, is equivalent and mimics the functionality of the native protein in in vitro growth inhibition assay experiments. We conclude that in the case of recombinant AMA1, the E. coli- and P. pastoris-derived antigens are immunologically and functionally equivalent and are unaffected by the posttranslational modification resulting from expression in these two systems.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Processamento de Proteína Pós-Traducional , Proteínas de Protozoários/imunologia , Vacinas Sintéticas/imunologia , Sequência de Aminoácidos , Animais , Escherichia coli/genética , Dados de Sequência Molecular , Pichia/genética , Coelhos , Proteínas Recombinantes/imunologia
8.
Vaccine ; 23(19): 2530-9, 2005 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-15752840

RESUMO

Montanide ISA 720 is an experimental adjuvant, formulated as water-in-oil emulsions, that induces high antibody titers in several animal species. It has been used in human vaccine trials with malaria and HIV vaccines. The heightened response is likely due, in part, to the formation of a depot at the injection site. However, post-formulation modifications were seen with seven proteins tested during storage of ISA 720 formulations at 37 degrees C for 1 week and two proteins stored longer at 4 degrees C. Potency studies in mice, in which the stored vaccines were diluted into placebo emulsions for appropriate dosing, indicated that this instability could lead to loss of immunogenicity in the post-injection depot, limiting the allowable storage time of preformed vaccines. We describe point-of-injection formulation for ISA 720 vaccines that meets the requirement for in vitro stability. For preformed vaccines, addition of glycine or glycylglycine prevented antigen modification on storage at 37 degrees C, providing a potential way of stabilizing antigen/ISA 720 formulations for in vitro storage and the post-injection depot.


Assuntos
Adjuvantes Imunológicos/normas , Antígenos/imunologia , Manitol/análogos & derivados , Ácidos Oleicos/normas , Vacinas/química , Vacinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antiprotozoários/sangue , Antígenos/administração & dosagem , Antígenos/química , Antígenos de Protozoários/imunologia , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Emulsões , Ensaio de Imunoadsorção Enzimática , Feminino , Manitol/administração & dosagem , Manitol/imunologia , Manitol/normas , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Ácidos Oleicos/administração & dosagem , Ácidos Oleicos/imunologia , Controle de Qualidade , Proteínas Recombinantes , Temperatura , Vacinas/administração & dosagem
9.
Vaccine ; 23(24): 3131-8, 2005 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-15837212

RESUMO

Plasmodium vivax is responsible for the majority of malaria cases outside of Africa, and results in substantial morbidity. Transmission blocking vaccines are a potentially powerful component of a multi-faceted public health approach to controlling or eliminating malaria. We report the first phase 1 clinical trial of a P. vivax transmission blocking vaccine in humans. The Pvs25H vaccine is a recombinant protein derived from the Pvs25 surface antigen of P. vivax ookinetes. The protein was expressed in Saccharomyces cerevisiae, purified, and adsorbed onto Alhydrogel. Ten volunteers in each of three dose groups (5, 20, or 80 microg) were vaccinated by intramuscular injection in an open-label study at 0, 28 and 180 days. No vaccine-related serious adverse events were observed. The majority of adverse events causally related to vaccination were mild or moderate in severity. Injection site tenderness was the most commonly observed adverse event. Anti-Pvs25H antibody levels measured by ELISA peaked after the third vaccination. Vaccine-induced antibody is functionally active as evidenced by significant transmission blocking activity in the membrane feeding assay. Correlation between antibody concentration and degree of inhibition was observed. Pvs25H generates transmission blocking immunity in humans against P. vivax demonstrating the potential of this antigen as a component of a transmission blocking vaccine.


Assuntos
Antígenos de Protozoários/uso terapêutico , Antígenos de Superfície/uso terapêutico , Vacinas Antimaláricas/uso terapêutico , Malária Vivax/prevenção & controle , Malária Vivax/transmissão , Adolescente , Adulto , Animais , Antígenos de Protozoários/efeitos adversos , Antígenos de Superfície/efeitos adversos , Culicidae/imunologia , Culicidae/parasitologia , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Esquemas de Imunização , Vacinas Antimaláricas/efeitos adversos , Masculino , Pessoa de Meia-Idade , Dinâmica não Linear , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/uso terapêutico
10.
Infect Immun ; 73(6): 3677-85, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15908397

RESUMO

Apical membrane antigen 1 (AMA1), a polymorphic merozoite surface protein, is a leading blood-stage malaria vaccine candidate. A phase 1 trial was conducted with 30 malaria-naive volunteers to assess the safety and immunogenicity of the AMA1-C1 malaria vaccine. AMA1-C1 contains an equal mixture of recombinant proteins based on sequences from the FVO and 3D7 clones of Plasmodium falciparum. The proteins were expressed in Pichia pastoris and adsorbed on Alhydrogel. Ten volunteers in each of three dose groups (5 mug, 20 mug, and 80 mug) were vaccinated in an open-label study at 0, 28, and 180 days. The vaccine was well tolerated, with pain at the injection site being the most commonly observed reaction. Anti-AMA1 immunoglobulin G (IgG) was detected by enzyme-linked immunosorbent assay (ELISA) in 15/28 (54%) volunteers after the second immunization and in 23/25 (92%) after the third immunization, with equal reactivity to both AMA1-FVO and AMA1-3D7 vaccine components. A significant dose-response relationship between antigen dose and antibody response by ELISA was observed, and the antibodies were predominantly of the IgG1 isotype. Confocal microscopic evaluation of sera from vaccinated volunteers demonstrated reactivity with P. falciparum schizonts in a pattern similar to native parasite AMA1. Antigen-specific in vitro inhibition of both FVO and 3D7 parasites was achieved with IgG purified from sera of vaccinees, demonstrating biological activity of the antibodies. To our knowledge, this is the first AMA1 vaccine candidate to elicit functional immune responses in malaria-naive humans, and our results support the further development of this vaccine.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Vacinas Sintéticas/imunologia , Adulto , Animais , Anticorpos Antiprotozoários/sangue , Feminino , Humanos , Imunoglobulina G/sangue , Vacinas Antimaláricas/efeitos adversos , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Plasmodium falciparum/crescimento & desenvolvimento
11.
Vaccine ; 21(15): 1650-7, 2003 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-12639486

RESUMO

In previously published studies, Saccharomyces cerevisiae recombinant protein expression systems have been employed to express the malaria parasite antigen Pfs25, a candidate transmission-blocking vaccine antigen against Plasmodium falciparum malaria. However, despite having been in two Phase 1 trials, the recombinant Pfs25 so produced (previously called TBV25H) exists as a mixture of two monomeric protein conformational forms, Pfs25H-A and Pfs25H-B. In this study, we optimized the expression and purification of the two Pfs25H conformers in S. cerevisiae, and characterized their biochemical and antigenic properties, immunogenicities, and transmission-blocking activities. Pfs25H-A is apparently homogeneous, and has the correct conformation as measured by monoclonal antibody recognition. It is, however, expressed at a low yield of only 0.19mg/l. By contrast, Pfs25H-B is produced as a heterogeneous population of molecules that do not seem to have the correct conformation. Nonetheless, both forms appear equally effective in their ability to produce transmission-blocking antibodies in mice. To address the low yield seen with S. cerevisiae, we also expressed Pfs25 in Pichia pastoris. P. pastoris is apparently superior to S. cerevisiae in producing higher yield, immunologically more potent, biologically more active Pfs25H-A.


Assuntos
Ensaios Clínicos como Assunto , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Pichia/metabolismo , Proteínas de Protozoários/biossíntese , Sequência de Aminoácidos , Animais , Ensaios Clínicos como Assunto/métodos , Culicidae/imunologia , Culicidae/parasitologia , Feminino , Regulação Fúngica da Expressão Gênica/imunologia , Humanos , Vacinas Antimaláricas/biossíntese , Vacinas Antimaláricas/imunologia , Malária Falciparum/transmissão , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Pichia/imunologia , Plasmídeos/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/isolamento & purificação , Saccharomyces cerevisiae/imunologia , Saccharomyces cerevisiae/isolamento & purificação , Saccharomyces cerevisiae/metabolismo , Vacinas Sintéticas/biossíntese , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
12.
Infect Immun ; 70(2): 692-701, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11796601

RESUMO

Vaccination of mice with yeast-secreted Plasmodium yoelii-derived 19-kilodalton merozoite surface protein 1 (yMSP1(19)) has been shown to afford protection from challenge with a lethal strain of P. yoelii. Sterile immunity can be achieved when MSP1(19) is emulsified in Freund adjuvant but not when it is adsorbed to aluminum hydroxide gel (alum). Because complete Freund adjuvant is not an acceptable adjuvant for use in humans, alternative adjuvants must be identified for formulating MSP1(19) as a vaccine for use in humans. To determine whether oligodeoxynucleotides with CpG motifs (ODN), reported to be a powerful new class of adjuvants, could enhance the immunogenicity of yMSP1(19), C57BL/6 mice were vaccinated either with yMSP1(19) formulated with Freund adjuvant, with alum, or with ODN plus alum and challenged intravenously with P. yoelii 17XL asexual blood-stage parasites. Adsorption of immunogen and adjuvant to alum was optimized by adjusting buffer (phosphate versus acetate) and pH. We found that the adjuvant combination of ODN plus alum with yMSP1(19), injected intraperitoneally (i.p.), increased immunoglobulin G (IgG) yMSP1(19)-specific antibody production 12-fold over Freund adjuvant given i.p., 3-fold over Freund adjuvant given subcutaneously (s.c.), 300-fold over alum given i.p., and 48-fold over alum given s.c. The predominant antibody isotype in the group receiving alum-ODN-yMSP1(19) was IgG1. Increased antibody levels correlated to protection from a challenge with P. yoelii 17XL. Supernatant cytokine levels of gamma interferon in yMSP1(19)-stimulated splenocytes were dramatically elevated in the alum-ODN-yMSP1(19) group. Interleukin-10 (IL-10) levels were also elevated; however, no IL-5 was detected. The cytokine profile, as well as the predominant IgG1 antibody isotype, suggests the protective immune response was a mixed Th1/Th2 response.


Assuntos
Adjuvantes Imunológicos , Hidróxido de Alumínio/imunologia , Ilhas de CpG/imunologia , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Proteína 1 de Superfície de Merozoito/imunologia , Oligodesoxirribonucleotídeos/imunologia , Plasmodium yoelii/imunologia , Vacinas Sintéticas/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Modelos Animais de Doenças , Géis , Imunoglobulina G/sangue , Isotipos de Imunoglobulinas/sangue , Malária/sangue , Malária/imunologia , Malária/parasitologia , Proteína 1 de Superfície de Merozoito/genética , Camundongos , Camundongos Endogâmicos C57BL , Recombinação Genética , Células Th1/imunologia , Células Th2/imunologia , Virulência
13.
Protein Expr Purif ; 25(1): 87-96, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12071703

RESUMO

The budding yeast Saccharomyces cerevisiae has been used to express the recombinant protein Pvs25H, currently the only candidate transmission-blocking vaccine against Plasmodium vivax malaria. This molecule contains four epidermal growth factor-like domains and is expressed as at least two stable monomeric forms with different physicochemical properties. Pvs25H-A is apparently homogeneous and seems to have a correct disulfide bond structure. By contrast, Pvs25H-B is produced as a heterogeneous population of molecules, some of which are associated with an as yet unidentified chromophore, and it contains both internal and N-terminal cleavages. We report here a procedure for successfully separating these two forms with a process suitable for clinical production of this antigen.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/isolamento & purificação , Antígenos de Superfície/química , Antígenos de Superfície/isolamento & purificação , Antígenos/química , Antígenos/isolamento & purificação , Vacinas Antimaláricas/química , Vacinas Antimaláricas/isolamento & purificação , Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/imunologia , Western Blotting , Cromatografia , Cromatografia Líquida de Alta Pressão , Ensaios Clínicos como Assunto , Dissulfetos , Fermentação , Humanos , Malária/prevenção & controle , Dados de Sequência Molecular , Plasmodium vivax/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo
14.
Infect Immun ; 70(11): 6013-20, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12379677

RESUMO

Immunity induced by the 19-kDa fragment of Plasmodium yoelii merozoite surface protein 1 (MSP1(19)) is dependent on high titers of specific antibodies present at the time of challenge and a continuing active immune response postinfection. However, the specificity of the active immune response postinfection has not been defined. In particular, it is not known whether anti-MSP1(19) antibodies that arise following infection alone are sufficient for protection. We developed systems to investigate whether an MSP1(19)-specific antibody response alone both prechallenge and postchallenge is sufficient for protection. We were able to exclude antibodies with other specificities, as well as any contribution of MSP1(19)-specific CD4(+) T cells acting independent of antibody, and we concluded that an immune response focused solely on MSP1(19)-specific antibodies is sufficient for protection. The data imply that the ability of natural infection to boost an MSP1(19)-specific antibody response should greatly improve vaccine efficacy.


Assuntos
Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Proteína 1 de Superfície de Merozoito/imunologia , Fragmentos de Peptídeos/imunologia , Plasmodium yoelii/imunologia , Transferência Adotiva , Animais , Especificidade de Anticorpos , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Vacinação
15.
Infect Immun ; 70(12): 6606-13, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438332

RESUMO

It is widely believed that subunit vaccines composed of multiple components will offer greater protection against challenge by malaria, and yet there is little experimental evidence to support this view. We set out to test this proposition in the Plasmodium yoelii challenge system in rodents by comparing the degree of protection conferred by immunization with a mixture of merozoite surface proteins to that conferred by single proteins. We therefore examined a defined protein mixture made of the epidermal growth factor-like domains of P. yoelli merozoite surface protein 1 (MSP1) and MSP4/5, the homologue of P. falciparum MSP4 and MSP5. In the present study we demonstrate that this combination of recombinant proteins dramatically enhances protection against lethal malaria challenge compared to either protein administered alone. Many mice immunized with the MSP4/5 plus MSP1(19) combination did not develop detectable parasitemia after challenge. Combined immunization with MSP1(19) and yMSP4/5, a product characterized by lower protective efficacy, also greatly enhanced protection by reducing peak parasitemias and increasing the numbers of survivors. In some combination trials, levels of antibodies to MSP1(19) were elevated compared to the MSP1(19) alone group; however, improved protection occurred regardless of whether boosting of the anti-MSP1(19) response was observed. Boosting of anti-MSP1(19) did not appear to be due to contaminating endotoxin in the EcMSP4/5 material since enhanced protection was observed in C3H/HeJ mice, which are endotoxin insensitive. Collectively, these experiments show that multiantigen combinations offer enhanced levels of protection against asexual stage infection and suggest that combinations of MSP1, MSP4, and MSP5 should be evaluated further for use in humans.


Assuntos
Antígenos de Protozoários/imunologia , Malária/prevenção & controle , Plasmodium yoelii/imunologia , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/genética , Feminino , Imunização , Malária/parasitologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
16.
J Biol Chem ; 278(9): 7264-9, 2003 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-12493733

RESUMO

The protozoan parasite Plasmodium causes malaria, with hundreds of millions of cases recorded annually. Protection against malaria infection can be conferred by antibodies against merozoite surface protein (MSP)-1, making it an attractive vaccine candidate. Here we present the structure of the C-terminal domains of MSP-1 (known as MSP-1(19)) from Plasmodium knowlesi. The structure reveals two tightly packed epidermal growth factor-like domains oriented head to tail. In domain 1, the molecule displays a histidine binding site formed primarily by a highly conserved tryptophan. The protein carries a pronounced overall negative charge primarily due to the large number of acidic groups in domain 2. To map protein binding surfaces on MSP-1(19), we have analyzed the crystal contacts in five different crystal environments, revealing that domain 1 is highly preferred in protein-protein interactions. A comparison of MSP-1(19) structures from P. knowlesi, P. cynomolgi, and P. falciparum shows that, although the overall protein folds are similar, the molecules show significant differences in charge distribution. We propose the histidine binding site in domain 1 as a target for inhibitors of protein binding to MSP-1, which might prevent invasion of the merozoite into red blood cells.


Assuntos
Proteína 1 de Superfície de Merozoito/química , Plasmodium knowlesi/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , DNA/metabolismo , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/metabolismo , Eritrócitos/metabolismo , Histidina/química , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Triptofano/química
17.
Infect Immun ; 72(1): 584-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14688140

RESUMO

Antibodies directed against Pfs25, a protein present on the surface of zygotes and ookinetes of Plasmodium falciparum, completely block pathogen transmission. We evaluated the immunomodulatory effect of CpG oligodeoxynucleotides (ODN) on the immunogenicity of recombinant Pfs25 (rPfs25) formulated in alum (Al). Immunization of mice with rPfs25 plus CpG ODN improved both the antibody titer (a 30-fold-higher antibody response than that with rPfs25-Al alone) and avidity. Coadministration of CpG ODN dramatically enhanced the titer of immunoglobulin G2A (IgG2a) compared to the titer of the IgG1-dominant response caused by rPfs25-Al alone, and the sera from the CpG ODN-coadministered group completely blocked the transmission of P. falciparum parasites to mosquitoes, as determined by membrane feeding assays. However, transmission-blocking experiments revealed that blocking efficacy was dependent on high-titer antibody levels, independent of isotypes. These results suggest that CpG ODN can be used as an adjuvant to enhance the immunogenicity of rPfs25 as a malaria transmission-blocking vaccine.


Assuntos
Adjuvantes Imunológicos , Vacinas Antimaláricas/imunologia , Malária Falciparum/transmissão , Oligodesoxirribonucleotídeos/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Compostos de Alúmen , Animais , Anticorpos Antiprotozoários/sangue , Afinidade de Anticorpos , Imunização , Imunização Secundária , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/administração & dosagem , Proteínas de Protozoários/genética
18.
Infect Immun ; 70(12): 6961-7, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438375

RESUMO

A major challenge facing malaria vaccine development programs is identifying efficacious combinations of antigens. To date, merozoite surface protein 1 (MSP1) is regarded as the leading asexual vaccine candidate. Apical membrane antigen 1 (AMA1) has been identified as another leading candidate for an asexual malaria vaccine, but without any direct in vivo evidence that a recombinant form of Plasmodium falciparum AMA1 would have efficacy. We evaluated the efficacy of a form of P. falciparum AMA1, produced in Pichia pastoris, by vaccinating Aotus vociferans monkeys and then challenging them with P. falciparum parasites. Significant protection from this otherwise lethal challenge with P. falciparum was observed. Five of six animals had delayed patency; two of these remained subpatent for the course of the infection, and two controlled parasite growth at <0.75% of red blood cells parasitized. The protection induced by AMA1 was superior to that obtained with a form of MSP1 used in the same trial. The protection induced by a combination vaccine of AMA1 and MSP1 was not superior to the protection obtained with AMA1 alone, although the immunity generated appeared to operate against both vaccine components.


Assuntos
Antígenos de Protozoários , Eritrócitos/parasitologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/imunologia , Animais , Aotidae , Vacinas Antimaláricas/administração & dosagem , Proteínas de Membrana/genética , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/imunologia , Parasitemia/prevenção & controle , Proteínas de Protozoários/genética , Proteínas Recombinantes/genética , Vacinação , Vacinas Combinadas , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
19.
Infect Immun ; 71(12): 6766-74, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14638762

RESUMO

Protection against Plasmodium falciparum can be induced by vaccination in animal models with merozoite surface protein 1 (MSP1), which makes this protein an attractive vaccine candidate for malaria. In an attempt to produce a product that is easily scaleable and inexpensive, we expressed the C-terminal 42 kDa of MSP1 (MSP1(42)) in Escherichia coli, refolded the protein to its native form from insoluble inclusion bodies, and tested its ability to elicit antibodies with in vitro and in vivo activities. Biochemical, biophysical, and immunological characterization confirmed that refolded E. coli MSP1(42) was homogeneous and highly immunogenic. In a formulation suitable for human use, rabbit antibodies were raised against refolded E. coli MSP1(42) and tested in vitro in a P. falciparum growth invasion assay. The antibodies inhibited the growth of parasites expressing either homologous or heterologous forms of P. falciparum MSP1(42). However, the inhibitory activity was primarily a consequence of antibodies directed against the C- terminal 19 kDa of MSP1 (MSP1(19)). Vaccination of nonhuman primates with E. coli MSP1(42) in Freund's adjuvant protected six of seven Aotus monkeys from virulent infection with P. falciparum. The protection correlated with antibody-dependent mechanisms. Thus, this new construct, E. coli MSP1(42), is a viable candidate for human vaccine trials.


Assuntos
Escherichia coli/metabolismo , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Proteína 1 de Superfície de Merozoito/química , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium falciparum/imunologia , Dobramento de Proteína , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Aotus trivirgatus , Eritrócitos/imunologia , Eritrócitos/parasitologia , Escherichia coli/genética , Humanos , Vacinas Antimaláricas/administração & dosagem , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/metabolismo , Dados de Sequência Molecular , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação
20.
Proc Natl Acad Sci U S A ; 99(6): 3860-5, 2002 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11904437

RESUMO

Immunity to Plasmodium falciparum in African children has been correlated with antibodies to the P. falciparum erythrocyte membrane protein 1 (PfEMP1) variant gene family expressed on the surface of infected red cells. We immunized Aotus monkeys with a subregion of the Malayan Camp variant antigen (MCvar1) that mediates adhesion to the host receptor CD36 on the endothelial surface and present data that PfEMP1 is an important target for vaccine development. The immunization induced a high level of protection against the homologous strain. Protection correlated with the titer of agglutinating antibodies and occurred despite the expression of variant copies of the gene during recurrent waves of parasitemia. A second challenge with a different P. falciparum strain, to which there was no agglutinating activity, showed no protection but boosted the immune response to this region during the infection. The level of protection and the evidence of boosting during infection encourage further exploration of this concept for malaria vaccine development.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Aotus trivirgatus/imunologia , Aotus trivirgatus/parasitologia , Variação Genética/genética , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Vacinas Antimaláricas/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Masculino , Mutação/genética , Parasitemia/imunologia , Parasitemia/parasitologia , Parasitemia/prevenção & controle , Plasmodium falciparum/classificação , Plasmodium falciparum/genética , Estrutura Terciária de Proteína , Vacinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA