Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 4: 176, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23847617

RESUMO

Genetically inactivated, Gram-negative bacteria that express malaria vaccine candidates represent a promising novel self-adjuvanting vaccine approach. Antigens expressed on particulate bacterial carriers not only target directly to antigen-presenting cells but also provide a strong danger signal thus circumventing the requirement for potent extraneous adjuvants. E. coli expressing malarial antigens resulted in the induction of either Th1 or Th2 biased responses that were dependent on both antigen and sub-cellular localization. Some of these constructs induced higher quality humoral responses compared to recombinant protein and most importantly they were able to induce sterile protection against sporozoite challenge in a murine model of malaria. In light of these encouraging results, two major Plasmodium falciparum pre-erythrocytic malaria vaccine targets, the Cell-Traversal protein for Ookinetes and Sporozoites (CelTOS) fused to the Maltose-binding protein in the periplasmic space and the Circumsporozoite Protein (CSP) fused to the Outer membrane (OM) protein A in the OM were expressed in a clinically relevant, attenuated Shigella strain (Shigella flexneri 2a). This type of live-attenuated vector has previously undergone clinical investigations as a vaccine against shigellosis. Using this novel delivery platform for malaria, we find that vaccination with the whole-organism represents an effective vaccination alternative that induces protective efficacy against sporozoite challenge. Shigella GeMI-Vax expressing malaria targets warrant further evaluation to determine their full potential as a dual disease, multivalent, self-adjuvanting vaccine system, against both shigellosis, and malaria.

2.
Vaccine ; 29(35): 5940-9, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21722682

RESUMO

The malarial protein CelTOS, for cell-traversal protein for ookinetes and sporozoites, from Plasmodium berghei has been shown to mediate malarial invasion of both vertebrate and insect host cells and is required for establishing their successful infections. In the vertebrate host, Plasmodium sporozoites traverse via a complex passage through cellular barriers in the skin and the liver sinusoid to infect hepatocytes. Induction of immunity targeted to molecules involved in sporozoite motility and migration into hepatocytes may lead to abrogation of hepatocyte infection. We have previously demonstrated the potential of CelTOS as a target antigen for a pre-erythrocytic vaccine. The objective of the current study was to determine the potency of different vaccine platforms to induce protective immunity and determine the mode of action in protective immune responses. To this end, inbred Balb/c and outbred ICR mice were immunized with either the recombinant protein adjuvanted with Montanide ISA-720 or with a pCI-TPA plasmid encoding the P. berghei CelTOS (epidermal delivery by gene-gun) and assessed for the induction of protective responses against a homologous P. berghei challenge. Humoral and cellular immune responses induced by the various immunization regimens were evaluated in an effort to establish immune correlates. The results confirm that the CelTOS antigen is a potentially interesting pre-erythrocytic vaccine candidate and demonstrate that both arms of the adaptive immune system are required to mediate complete sterile protection against sporozoite challenge.


Assuntos
Anticorpos Antiprotozoários/sangue , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Proteínas de Protozoários/imunologia , Esporozoítos/fisiologia , Linfócitos T/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Biolística , Dicroísmo Circular , Soros Imunes/imunologia , Malária/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética , Coelhos , Esporozoítos/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia
3.
PLoS One ; 5(8): e12294, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20808868

RESUMO

BACKGROUND: The Plasmodium protein Cell-traversal protein for ookinetes and sporozoites (CelTOS) plays an important role in cell traversal of host cells in both, mosquito and vertebrates, and is required for successful malaria infections. CelTOS is highly conserved among the Plasmodium species, suggesting an important functional role across all species. Therefore, targeting the immune response to this highly conserved protein and thus potentially interfering with its biological function may result in protection against infection even by heterologous species of Plasmodium. METHODOLOGY/PRINCIPAL FINDINGS: To test this hypothesis, we developed a recombinant codon-harmonized P. falciparum CelTOS protein that can be produced to high yields in the E. coli expression system. Inbred Balb/c and outbred CD-1 mice were immunized with various doses of the recombinant protein adjuvanted with Montanide ISA 720 and characterized using in vitro and in vivo analyses. CONCLUSIONS/SIGNIFICANCE: Immunization with PfCelTOS resulted in potent humoral and cellular immune responses and most importantly induced sterile protection against a heterologous challenge with P. berghei sporozoites in a proportion of both inbred and outbred mice. The biological activity of CelTOS-specific antibodies against the malaria parasite is likely linked to the impairment of sporozoite motility and hepatocyte infectivity. The results underscore the potential of this antigen as a pre-erythrocytic vaccine candidate and demonstrate for the first time a malaria vaccine that is cross-protective between species.


Assuntos
Antígenos de Protozoários/imunologia , Eritrócitos , Imunização/métodos , Plasmodium berghei/imunologia , Plasmodium falciparum/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Reações Cruzadas , Escherichia coli/genética , Feminino , Hepatócitos/parasitologia , Humanos , Interferon gama/biossíntese , Interleucina-4/biossíntese , Manitol/análogos & derivados , Camundongos , Movimento , Ácidos Oleicos , Plasmodium falciparum/fisiologia , Solubilidade , Esporozoítos/imunologia , Esporozoítos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
4.
Vaccine ; 27(41): 5700-8, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19576940

RESUMO

Malaria represents a major global health problem but despite extensive efforts, no effective vaccine is available. Various vaccine candidates have been developed that provide protection in animal models, such as a gene gun-delivered DNA vaccine encoding the circumsporozoite protein (CSP) of Plasmodium berghei. A common shortcoming of most malaria vaccines is the requirement for multiple immunizations leaving room for improvement even for established vaccine candidates such as the CSP-DNA vaccine. In this study, we explored whether regulating apoptosis in DNA vaccine transfected host cells could accelerate the onset of protective immunity and provide significant protection after a single immunization. A pro-apoptotic gene (Bax) was used as a molecular adjuvant in an attempt to mimic the immunostimulatory apoptosis triggered by viral or virus-derived vaccines, while anti-apoptotic genes such as Bcl-XL may increase the life span of transfected cells thus prolonging antigen production. Surprisingly, co-delivery of either Bax or Bcl-XL greatly reduced CSP-DNA vaccine efficacy after a single immunization. Co-delivery of Bax for three immunizations still had a detrimental effect on protective immunity, while repeated co-delivery of Bcl-XL had no negative impact. The fine characterization of humoral and cellular immune response modulated by these two molecular adjuvants revealed a previously unknown effect, i.e., a shift in the Th-profile. These results demonstrate that pro- or anti-apoptotic molecules should not be used as molecular adjuvants without careful evaluation of the resulting immune response. This finding represents yet another example that strategies to enhance vaccine efficacy developed for other model systems such as viral diseases cannot easily be applied to any vaccine.


Assuntos
Adjuvantes Imunológicos/farmacologia , Apoptose , Vacinas Antimaláricas/imunologia , Plasmodium berghei/imunologia , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Proteína X Associada a bcl-2/farmacologia , Proteína bcl-X/farmacologia , Adjuvantes Imunológicos/genética , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/genética , Proteína X Associada a bcl-2/genética , Proteína bcl-X/genética
5.
Vaccine ; 25(45): 7732-6, 2007 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-17931754

RESUMO

Immune response against circumsporozoite protein (CSP) of Plasmodium berghei, a major surface protein on the sporozoite, confers protection in various murine malaria models. Engineered DNA vaccine encoding CSP and 3 copies of C3d caused an unexpected loss in protection attributed to the binding of C3d to the C-terminal region of CSP. Because the C3d region known as p28 represents the complement receptor (CR) 2-binding motif, we developed a CSP-3 copies of p28 DNA construct (CSP-3p28). CSP-3p28-immunized mice were better protected against P. berghei sporozoites than CSP-immunized mice 6 weeks after the 2nd boost, produced sufficient IgG1 anti-CSP and CSP C-terminus antibody and failed to produce IgG2a. CSP-3C3d-immunized mice were not protected, failed to produce IgG1 and produced high amounts of IgG2a. We conclude that use of the CR2-binding motif of C3d as molecular adjuvant to CSP results in anti-malaria protective immune response probably by targeting the chimeric protein to CR2.


Assuntos
Complemento C3d/metabolismo , Malária/imunologia , Fragmentos de Peptídeos/administração & dosagem , Plasmodium berghei/imunologia , Proteínas de Protozoários/imunologia , Proteínas Recombinantes de Fusão/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antiprotozoários/imunologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/imunologia , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/uso terapêutico , Esporozoítos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...