Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(5): e36245, 2012.
Article in English | MEDLINE | ID: mdl-22567144

ABSTRACT

Immunological adjuvants that induce T cell-mediate immunity (TCMI) with the least side effects are needed for the development of human vaccines. Glycoinositolphospholipids (GIPL) and CpGs oligodeoxynucleotides (CpG ODNs) derived from the protozoa parasite Trypanosoma cruzi induce potent pro-inflammatory reaction through activation of Toll-Like Receptor (TLR)4 and TLR9, respectively. Here, using mouse models, we tested the T. cruzi derived TLR agonists as immunological adjuvants in an antitumor vaccine. For comparison, we used well-established TLR agonists, such as the bacterial derived monophosphoryl lipid A (MPL), lipopeptide (Pam3Cys), and CpG ODN. All tested TLR agonists were comparable to induce antibody responses, whereas significant differences were noticed in their ability to elicit CD4(+) T and CD8(+) T cell responses. In particular, both GIPLs (GTH, and GY) and CpG ODNs (B344, B297 and B128) derived from T. cruzi elicited interferon-gamma (IFN-γ) production by CD4(+) T cells. On the other hand, the parasite derived CpG ODNs, but not GIPLs, elicited a potent IFN-γ response by CD8(+) T lymphocytes. The side effects were also evaluated by local pain (hypernociception). The intensity of hypernociception induced by vaccination was alleviated by administration of an analgesic drug without affecting protective immunity. Finally, the level of protective immunity against the NY-ESO-1 expressing melanoma was associated with the magnitude of both CD4(+) T and CD8(+) T cell responses elicited by a specific immunological adjuvant.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Immunity, Cellular/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Trypanosoma cruzi/immunology , Adjuvants, Immunologic , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Melanoma/immunology , Melanoma/metabolism , Mice
2.
PLoS Negl Trop Dis ; 5(9): e1310, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21931874

ABSTRACT

BACKGROUND: Visceral leishmaniasis is the most severe form of leishmaniasis. Approximately 20% of zoonotic human visceral leishmaniasis worldwide is caused by Leishmania infantum, which is also known as Leishmania chagasi in Latin America, and disease incidence is increasing in urban and peri-urban areas of the tropics. In this form of disease, dogs are the main reservoirs. Diagnostic methods used to identify Leishmania infected animals are not able to detect all of the infected ones, which can compromise the effectiveness of disease control. Therefore, to contribute to the improvement of diagnostic methods for canine visceral leishmaniasis (CVL), we aimed to identify and test novel antigens using high-throughput analysis. METHODOLOGY/PRINCIPAL FINDINGS: Immunodominant proteins from L. infantum were mapped in silico to predict B cell epitopes, and the 360 predicted peptides were synthesized on cellulose membranes. Immunoassays were used to select the most reactive peptides, which were then investigated with canine sera. Next, the 10 most reactive peptides were synthesized using solid phase peptide synthesis protocol and tested using ELISA. The sensitivity and specificity of these peptides were also compared to the EIE-LVC Bio-Manguinhos kit, which is recommended by the Brazilian Ministry of Health for use in leishmaniasis control programs. The sensitivity and specificity of the selected synthesized peptides was as high as 88.70% and 95.00%, respectively, whereas the EIE-LVC kit had a sensitivity of 13.08% and 100.00% of specificity. Although the tests based on synthetic peptides were able to diagnose up to 94.80% of asymptomatic dogs with leishmaniasis, the EIE-LVC kit failed to detect the disease in any of the infected asymptomatic dogs. CONCLUSIONS/SIGNIFICANCE: Our study shows that ELISA using synthetic peptides is a technique with great potential for diagnosing CVL; furthermore, the use of these peptides in other diagnostic methodologies, such as immunochromatographic tests, could be beneficial to CVL control programs.


Subject(s)
Antigens, Protozoan/analysis , Dog Diseases/parasitology , Enzyme-Linked Immunosorbent Assay/methods , High-Throughput Screening Assays/methods , Leishmaniasis, Visceral/veterinary , Peptides/analysis , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Asymptomatic Infections , Disease Reservoirs/parasitology , Disease Reservoirs/veterinary , Dog Diseases/diagnosis , Dogs , High-Throughput Screening Assays/standards , Leishmaniasis, Visceral/diagnosis , Peptides/chemical synthesis , Peptides/immunology , ROC Curve , Reproducibility of Results , Sensitivity and Specificity
3.
Vaccine ; 29(27): 4476-84, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21549794

ABSTRACT

Toxoplasma gondii is an intracellular parasite widely spread around the world. The surface antigens (SAG) 1, 2 and 3 are the main proteins expressed on the surface of T. gondii tachyzoites. Replication-defective adenovirus serotype 5 (rAd5) is one of the most potent recombinant viral vectors for eliciting T cell-mediated immunity in mice and humans. Here we show that vaccination with rAd5 expressing SAG1 (AdSAG1), but neither SAG2 nor SAG3, induces protective immunity in the highly susceptible C57BL/6 mice challenged with T. gondii. Furthermore, we evaluated different immunological components involved on viral induced protective immunity. We observed that host protection elicited by AdSAG1 is highly dependent on IL-12, IFN-γ and CD8(+) T lymphocytes. Importantly, the induction of protective immunity (T cell-derived IFN-γ) was also dependent on Myeloid Differentiation Factor 88 (MyD88), and thus, likely to involve Toll-like Receptors. We conclude that protective parasite specific-CD8(+) T cells are elicited by a mechanism that involves MyD88-dependent induction of IL-12.


Subject(s)
Adenoviridae/genetics , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Myeloid Differentiation Factor 88/metabolism , Protozoan Vaccines/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Adenoviridae/metabolism , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Antigens, Surface/genetics , Antigens, Surface/immunology , Antigens, Surface/metabolism , Female , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/genetics , Toxoplasma/genetics , Toxoplasma/metabolism , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/prevention & control
4.
Vaccine ; 26(35): 4585-93, 2008 Aug 18.
Article in English | MEDLINE | ID: mdl-18588933

ABSTRACT

A2 was identified as an amastigote virulence factor of Leishmania (Leishmania) donovani and as a candidate antigen for vaccine development against visceral leishmaniasis. Here, predicted hydrophilic, class I and II MHC-binding synthetic peptides were used to define epitopes recognized by A2-specific antibodies, CD8+ T and CD4+ T cells, respectively. Immunization of BALB/c mice with adenovirus expressing A2 (AdA2) resulted in low antibody response, contrasting with high levels of IFN-gamma producing CD4+ T and CD8+ T cells specific for A2. Further, A2-specific CD8+ T cells from immunized mice were capable of lysing sensitized target cells in vivo. Finally, we demonstrated an association of A2-specific T cell responses and reduced parasitism in both liver and spleen from mice immunized with AdA2 and challenged with L. (L.) chagasi.


Subject(s)
Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Epitope Mapping , Interferon-gamma/immunology , Leishmaniasis Vaccines/immunology , Protozoan Proteins/immunology , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity Tests, Immunologic , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Female , Genetic Vectors , Leishmania donovani/genetics , Leishmaniasis Vaccines/genetics , Liver/parasitology , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Protozoan Proteins/genetics , Spleen/parasitology
5.
J Immunol ; 180(3): 1737-48, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209071

ABSTRACT

Interference or competition between CD8(+) T cells restricted by distinct MHC-I molecules can be a powerful means to establish an immunodominant response. However, its importance during infections is still questionable. In this study, we describe that following infection of mice with the human pathogen Trypanosoma cruzi, an immunodominant CD8(+) T cell immune response is developed directed to an H-2K(b)-restricted epitope expressed by members of the trans-sialidase family of surface proteins. To determine whether this immunodominance was exerted over other non-H-2K(b)-restricted epitopes, we measured during infection of heterozygote mice, immune responses to three distinct epitopes, all expressed by members of the trans-sialidase family, recognized by H-2K(b)-, H-2K(k)-, or H-2K(d)-restricted CD8(+) T cells. Infected heterozygote or homozygote mice displayed comparably strong immune responses to the H-2K(b)-restricted immunodominant epitope. In contrast, H-2K(k)- or H-2K(d)-restricted immune responses were significantly impaired in heterozygote infected mice when compared with homozygote ones. This interference was not dependent on the dose of parasite or the timing of infection. Also, it was not seen in heterozygote mice immunized with recombinant adenoviruses expressing T. cruzi Ags. Finally, we observed that the immunodominance was circumvented by concomitant infection with two T. cruzi strains containing distinct immunodominant epitopes, suggesting that the operating mechanism most likely involves competition of T cells for limiting APCs. This type of interference never described during infection with a human parasite may represent a sophisticated strategy to restrict priming of CD8(+) T cells of distinct specificities, avoiding complete pathogen elimination by host effector cells, and thus favoring host parasitism.


Subject(s)
Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , H-2 Antigens/immunology , Immunodominant Epitopes/immunology , Trypanosoma cruzi/immunology , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , H-2 Antigens/chemistry , H-2 Antigens/genetics , Heterozygote , Immunization , Mice , Mice, Inbred Strains , Molecular Sequence Data
6.
Vaccine ; 25(32): 6018-27, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17629597

ABSTRACT

We recently described that vaccination of mice with a glutathione S transferase fusion protein representing amino acids 261-500 of the Amastigote Surface Protein-2 efficiently cross-primed protective CD8+ T cells against a lethal challenge with the human protozoan parasite Trypanosoma cruzi. In this study, we initially established that this protective immunity was long lived. Subsequently, we studied the importance of TLR9 agonist CpG ODN 1826, TLR4 and CD4+ T cells for the generation of these protective CD8+ T cells. We found that: (i) the TLR9 agonist CpG ODN 1826 improved the efficiency of protective immunity; (ii) TLR4 is not relevant for priming of specific CD8+ T cells; (iii) CD4+ T cells are critical for priming of memory/protective CD8+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Chagas Disease/prevention & control , Cross-Priming/immunology , Toll-Like Receptor 9/immunology , Trypanosoma cruzi/immunology , Adjuvants, Immunologic , Animals , DNA/immunology , Epitopes/immunology , Female , Mice , Mice, Inbred C3H , Neuraminidase/immunology , Oligodeoxyribonucleotides , Protozoan Vaccines/immunology , Time Factors , Toll-Like Receptor 9/agonists
7.
Hum Gene Ther ; 17(4): 415-26, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16610929

ABSTRACT

We have generated recombinant adenoviruses encoding three genetically modified surface antigens (SAGs) of the parasite Toxoplasma gondii, that is, AdSAG1, AdSAG2, and AdSAG3. Modifications included the removal of their glycosylphosphatidylinositol (GPI) anchoring motifs and, in some cases, the exchange of the native signal peptide for influenza virus hemagglutinin signal sequence. Adenovirus immunization of BALB/c mice elicited potent antibody responses against each protein, displaying a significant bias toward a helper T cell type 1 (Th1) profile in animals vaccinated with AdSAG1. Furthermore, the presence of parasite-specific IFN-gamma-producing T cells was analyzed by proliferation assays and enzyme-linked immunospot assays in the same animals. Splenocytes from immunized mice secreted IFN-gamma after in vitro stimulation with tachyzoite lysate antigen or with a fraction enriched for membrane-purified GPI-anchored proteins (F3) from the T. gondii tachyzoite surface. Epitopes recognized by CD8+ T cells were identified in SAG1 and SAG3, but not SAG2, sequences, although this protein also induced a specific response. We also tested the capacity of the immune responses detected to protect mice against a challenge with live T. gondii parasites. Although no protection was observed against tachyzoites of the highly virulent RH strain, a significant reduction in cyst loads in the brain was observed in animals challenged with the P-Br strain. Thus, up to 80% of the cysts were eliminated from animals vaccinated with a mixture of the three recombinant viruses. Because adenoviruses seemed capable of inducing Th1-biased protective immune responses against T. gondii antigens, other parasite antigens should be tested alone or in combination with those described here to further develop a protective vaccine against toxoplasmosis.


Subject(s)
Adenoviridae , Antigens, Protozoan/genetics , Membrane Glycoproteins/genetics , Protozoan Proteins/genetics , Protozoan Vaccines/genetics , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Antigens, Protozoan/immunology , Female , Gene Deletion , Immunity, Active , Immunity, Cellular , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Recombination, Genetic , Toxoplasmosis, Animal/prevention & control , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virus Replication/genetics
8.
Infect Immun ; 74(4): 2477-81, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16552083

ABSTRACT

The kinetics of effector CD8+-T-cell responses to specific Trypanosoma cruzi epitopes was investigated after challenge. Our results suggest that the delayed kinetics differs from that observed in other microbial infections and facilitates the establishment of the disease in naïve mice. In contrast, in vaccinated mice, the swift CD8+-T-cell response helps host survival after challenge.


Subject(s)
Chagas Disease/immunology , Cytotoxicity Tests, Immunologic , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/immunology , T-Lymphocytes, Cytotoxic/immunology , Trypanosoma cruzi/immunology , Animals , CD8 Antigens/genetics , Chagas Disease/mortality , Chagas Disease/prevention & control , Epitopes, T-Lymphocyte/immunology , Glycoproteins/immunology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/metabolism , Kinetics , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neuraminidase/immunology , Perforin , Pore Forming Cytotoxic Proteins , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/parasitology
9.
Infect Immun ; 73(9): 6017-25, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16113322

ABSTRACT

We previously described that DNA vaccination with the gene encoding amastigote surface protein 2 (ASP-2) protects approximately 65% of highly susceptible A/Sn mice against the lethal Trypanosoma cruzi infection. Here, we explored the possibility that bacterial recombinant proteins of ASP-2 could be used to improve the efficacy of vaccinations. Initially, we compared the protective efficacy of vaccination regimens using either a plasmid DNA, a recombinant protein, or both sequentially (DNA priming and protein boosting). Survival after the challenge was not statistically different among the three mouse groups and ranged from 53.5 to 75%. The fact that immunization with a recombinant protein alone induced protective immunity revealed the possibility that this strategy could be pursued for vaccination. We investigated this possibility by using six different recombinant proteins representing distinct portions of ASP-2. The vaccination of mice with glutathione S-transferase fusion proteins representing amino acids 261 to 500 or 261 to 380 of ASP-2 in the presence of the adjuvants alum and CpG oligodeoxynucleotide 1826 provided remarkable immunity, consistently protecting 100% of the A/Sn mice. Immunity was completely reversed by the in vivo depletion of CD8(+) T cells, but not CD4(+) T cells, and was associated with the presence of CD8(+) T cells specific for an epitope located between amino acids 320 and 327 of ASP-2. We concluded that a relatively simple formulation consisting of a recombinant protein with a selected portion of ASP-2, alum, and CpG oligodeoxynucleotide 1826 might be used to cross-prime strong CD8(+)-T-cell-dependent protective immunity against T. cruzi infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Neuraminidase/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Adjuvants, Immunologic/genetics , Animals , Chagas Disease/genetics , Chagas Disease/mortality , Chagas Disease/prevention & control , Disease Susceptibility , Female , Mice , Mice, Inbred A , Neuraminidase/administration & dosage , Neuraminidase/genetics , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/genetics , Trypanosoma cruzi/enzymology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...