Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Pharmacol Res ; 163: 105292, 2021 01.
Article in English | MEDLINE | ID: mdl-33171305

ABSTRACT

Resolution failure of exacerbated inflammation triggered by Influenza A virus (IAV) prevents return of pulmonary homeostasis and survival, especially when associated with secondary pneumococcal infection. Therapeutic strategies based on pro-resolving molecules have great potential against acute inflammatory diseases. Angiotensin-(1-7) [Ang-(1-7)] is a pro-resolving mediator that acts on its Mas receptor (MasR) to promote resolution of inflammation. We investigated the effects of Ang-(1-7) and the role of MasR in the context of primary IAV infection and secondary pneumococcal infection and evaluated pulmonary inflammation, virus titers and bacteria counts, and pulmonary damage. Therapeutic treatment with Ang-(1-7) decreased neutrophil recruitment, lung injury, viral load and morbidity after a primary IAV infection. Ang-(1-7) induced apoptosis of neutrophils and efferocytosis of these cells by alveolar macrophages, but had no direct effect on IAV replication in vitro. MasR-deficient (MasR-/-) mice were highly susceptible to IAV infection, displaying uncontrolled inflammation, increased viral load and greater lethality rate, as compared to WT animals. Ang-(1-7) was not protective in MasR-/- mice. Interestingly, Ang-(1-7) given during a sublethal dose of IAV infection greatly reduced morbidity associated with a subsequent S. pneumoniae infection, as seen by decrease in the magnitude of neutrophil influx, number of bacteria in the blood leading to a lower lethality. Altogether, these results show that Ang-(1-7) is highly protective against severe primary IAV infection and protects against secondary bacterial infection of the lung. These effects are MasR-dependent. Mediators of resolution of inflammation, such as Ang-(1-7), should be considered for the treatment of pulmonary viral infections.


Subject(s)
Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Peptide Fragments/therapeutic use , Pneumococcal Infections/drug therapy , Pneumonia, Viral/drug therapy , Proto-Oncogene Proteins/immunology , Receptors, G-Protein-Coupled/immunology , A549 Cells , Angiotensin I/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Dogs , Humans , Influenza A virus , Lung/drug effects , Lung/immunology , Lung/pathology , Madin Darby Canine Kidney Cells , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Peptide Fragments/pharmacology , Peroxidase/immunology , Phagocytosis/drug effects , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Streptococcus pneumoniae
2.
PLoS Pathog ; 12(4): e1005593, 2016 04.
Article in English | MEDLINE | ID: mdl-27128676

ABSTRACT

The ß1i, ß2i and ß5i immunoproteasome subunits have an important role in defining the repertoire of MHC class I-restricted epitopes. However, the impact of combined deficiency of the three immunoproteasome subunits in the development of protective immunity to intracellular pathogens has not been investigated. Here, we demonstrate that immunoproteasomes play a key role in host resistance and genetic vaccination-induced protection against the human pathogen Trypanosoma cruzi (the causative agent of Chagas disease), immunity to which is dependent on CD8+ T cells and IFN-γ (the classical immunoproteasome inducer). We observed that infection with T. cruzi triggers the transcription of immunoproteasome genes, both in mice and humans. Importantly, genetically vaccinated or T. cruzi-infected ß1i, ß2i and ß5i triple knockout (TKO) mice presented significantly lower frequencies and numbers of splenic CD8+ effector T cells (CD8+CD44highCD62Llow) specific for the previously characterized immunodominant (VNHRFTLV) H-2Kb-restricted T. cruzi epitope. Not only the quantity, but also the quality of parasite-specific CD8+ T cell responses was altered in TKO mice. Hence, the frequency of double-positive (IFN-γ+/TNF+) or single-positive (IFN-γ+) cells specific for the H-2Kb-restricted immunodominant as well as subdominant T. cruzi epitopes were higher in WT mice, whereas TNF single-positive cells prevailed among CD8+ T cells from TKO mice. Contrasting with their WT counterparts, TKO animals were also lethally susceptible to T. cruzi challenge, even after an otherwise protective vaccination with DNA and adenoviral vectors. We conclude that the immunoproteasome subunits are key determinants in host resistance to T. cruzi infection by influencing both the magnitude and quality of CD8+ T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Proteasome Endopeptidase Complex/immunology , Protozoan Vaccines/immunology , Adolescent , Adult , Animals , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Real-Time Polymerase Chain Reaction , Trypanosoma cruzi , Young Adult
3.
Hum Gene Ther ; 25(4): 350-63, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24568548

ABSTRACT

Heterologous prime-boost vaccination using plasmid DNA followed by replication-defective adenovirus vector generates a large number of specific CD8⁺ T effector memory (TEM) cells that provide long-term immunity against a variety of pathogens. In the present study, we initially characterized the frequency, phenotype, and function of these T cells in vaccinated mice that were subjected to infectious challenge with the human protozoan parasite Trypanosoma cruzi. We observed that the frequency of the specific CD8⁺ T cells in the spleens of the vaccinated mice increased after challenge. Specific TEM cells differentiated into cells with a KLRG1(High) CD27(Low) CD43(Low) CD183(Low)T-bet(High) Eomes(Low) phenotype and capable to produce simultaneously the antiparasitic mediators IFNγ and TNF. Using the gzmBCreERT2/ROSA26EYFP transgenic mouse line, in which the cells that express Granzyme B after immunization, are indelibly labeled with enhanced yellow fluorescent protein, we confirmed that CD8⁺ T cells present after challenge were indeed TEM cells that had been induced by vaccination. Subsequently, we observed that the in vivo increase in the frequency of the specific CD8⁺ T cells was not because of an anamnestic immune response. Most importantly, after challenge, the increase in the frequency of specific cells and the protective immunity they mediate were insensitive to treatment with the cytostatic toxic agent hydroxyurea. We have previously described that the administration of the drug FTY720, which reduces lymphocyte recirculation, severely impairs protective immunity, and our evidence supports the model that when large amounts of antigen-experienced CD8⁺ TEM cells are present after heterologous prime-boost vaccination, differentiation, and recirculation, rather than proliferation, are key for the resultant protective immunity.


Subject(s)
Adenoviridae/genetics , CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Genetic Vectors/genetics , Immunologic Memory , Trypanosoma cruzi/immunology , Adenoviridae/immunology , Animals , Animals, Genetically Modified , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chagas Disease/prevention & control , Disease Models, Animal , Female , Genetic Vectors/immunology , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Lymphocyte Count , Mice , Spleen/immunology , T-Cell Antigen Receptor Specificity/immunology , Vaccination , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
4.
PLoS One ; 8(5): e64443, 2013.
Article in English | MEDLINE | ID: mdl-23696894

ABSTRACT

Influenza virus A (IAV) causes annual epidemics and intermittent pandemics that affect millions of people worldwide. Potent inflammatory responses are commonly associated with severe cases of IAV infection. The complement system, an important mechanism of innate and humoral immune responses to infections, is activated during primary IAV infection and mediates, in association with natural IgM, viral neutralization by virion aggregation and coating of viral hemmagglutinin. Increased levels of the anaphylatoxin C5a were found in patients fatally infected with the most recent H1N1 pandemic virus. In this study, our aim was to evaluate whether targeting C5 activation alters inflammatory lung injury and viral load in a murine model of IAV infection. To address this question C57Bl/6j mice were infected intranasally with 10(4) PFU of the mouse adapted Influenza A virus A/WSN/33 (H1N1) or inoculated with PBS (Mock). We demonstrated that C5a is increased in bronchoalveolar lavage fluid (BALF) upon experimental IAV infection. To evaluate the role of C5, we used OmCI, a potent arthropod-derived inhibitor of C5 activation that binds to C5 and prevents release of C5a by complement. OmCI was given daily by intraperitoneal injection from the day of IAV infection until day 5. Treatment with OmCI only partially reduced C5a levels in BALF. However, there was significant inhibition of neutrophil and macrophage infiltration in the airways, Neutrophil Extracellular Traps (NETs) formation, death of leukocytes, lung epithelial injury and overall lung damage induced by the infection. There was no effect on viral load. Taken together, these data suggest that targeting C5 activation with OmCI during IAV infection could be a promising approach to reduce excessive inflammatory reactions associated with the severe forms of IAV infections.


Subject(s)
Complement C5/metabolism , Influenza A virus/pathogenicity , Lung Injury/etiology , Lung Injury/virology , Neutrophil Infiltration/physiology , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Animals , Humans , Influenza A virus/immunology , Male , Mice , Mice, Inbred C57BL
5.
PLoS Pathog ; 8(5): e1002699, 2012.
Article in English | MEDLINE | ID: mdl-22615561

ABSTRACT

MHC class Ia-restricted CD8(+) T cells are important mediators of the adaptive immune response against infections caused by intracellular microorganisms. Whereas antigen-specific effector CD8(+) T cells can clear infection caused by intracellular pathogens, in some circumstances, the immune response is suboptimal and the microorganisms survive, causing host death or chronic infection. Here, we explored the cellular and molecular mechanisms that could explain why CD8(+) T cell-mediated immunity during infection with the human protozoan parasite Trypanosoma cruzi is not optimal. For that purpose, we compared the CD8(+) T-cell mediated immune responses in mice infected with T. cruzi or vaccinated with a recombinant adenovirus expressing an immunodominant parasite antigen. Several functional and phenotypic characteristics of specific CD8(+) T cells overlapped. Among few exceptions was an accelerated expansion of the immune response in adenoviral vaccinated mice when compared to infected ones. Also, there was an upregulated expression of the apoptotic-signaling receptor CD95 on the surface of specific T cells from infected mice, which was not observed in the case of adenoviral-vaccinated mice. Most importantly, adenoviral vaccine provided at the time of infection significantly reduced the upregulation of CD95 expression and the proapoptotic phenotype of pathogen-specific CD8(+) cells expanded during infection. In parallel, infected adenovirus-vaccinated mice had a stronger CD8 T-cell mediated immune response and survived an otherwise lethal infection. We concluded that a suboptimal CD8(+) T-cell response is associated with an upregulation of CD95 expression and a proapoptotic phenotype. Both can be blocked by adenoviral vaccination.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neuraminidase/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , fas Receptor/biosynthesis , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Apoptosis , Chagas Disease/immunology , Chagas Disease/prevention & control , Interferon-gamma/biosynthesis , Lysosomal-Associated Membrane Protein 1/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Trypanosoma cruzi/pathogenicity , Vaccines, Synthetic/immunology
6.
Vaccine ; 30(18): 2882-91, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22381075

ABSTRACT

T-cell mediated immune responses are critical for acquired immunity against infection by the intracellular protozoan parasite Trypanosoma cruzi. Despite its importance, it is currently unknown where protective T cells are primed and whether they need to re-circulate in order to exert their anti-parasitic effector functions. Here, we show that after subcutaneous challenge, CD11c(+)-dependent specific CD8(+) T-cell immune response to immunodominant parasite epitopes arises almost simultaneously in the draining lymph node (LN) and the spleen. However, until day 10 after infection, we observed a clear upregulation of activation markers only on the surface of CD11C(+)PDCA1(+) cells present in the LN and not in the spleen. Therefore, we hypothesized that CD8(+) T cells re-circulated rapidly from the LN to the spleen. We investigated this phenomenon by administering FTY720 to T. cruzi-infected mice to prevent egress of T cells from the LN by interfering specifically with signalling through sphingosine-1-phosphate receptor-1. In T. cruzi-infected mice receiving FTY720, CD8 T-cell immune responses were higher in the draining LN and significantly reduced in their spleen. Most importantly, FTY720 increased susceptibility to infection, as indicated by elevated parasitemia and accelerated mortality. Similarly, administration of FTY720 to mice genetically vaccinated with an immunodominant parasite antigen significantly reduced their protective immunity, as observed by the parasitemia and survival of vaccinated mice. We concluded that re-circulation of lymphocytes mediated by sphingosine-1-phosphate receptor-1 greatly contributes to acquired and vaccine-induced protective immunity against experimental infection with a human protozoan parasite.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Receptors, Lysosphingolipid/metabolism , Trypanosoma cruzi/immunology , Animals , CD11c Antigen/analysis , CD8-Positive T-Lymphocytes/chemistry , Female , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Sphingosine-1-Phosphate Receptors , Spleen/immunology
7.
PLoS One ; 6(7): e22011, 2011.
Article in English | MEDLINE | ID: mdl-21779365

ABSTRACT

During adaptive immune response, pathogen-specific CD8(+) T cells recognize preferentially a small number of epitopes, a phenomenon known as immunodominance. Its biological implications during natural or vaccine-induced immune responses are still unclear. Earlier, we have shown that during experimental infection, the human intracellular pathogen Trypanosoma cruzi restricts the repertoire of CD8(+) T cells generating strong immunodominance. We hypothesized that this phenomenon could be a mechanism used by the parasite to reduce the breath and magnitude of the immune response, favoring parasitism, and thus that artificially broadening the T cell repertoire could favor the host. Here, we confirmed our previous observation by showing that CD8(+) T cells of H-2(a) infected mice recognized a single epitope of an immunodominant antigen of the trans-sialidase super-family. In sharp contrast, CD8(+) T cells from mice immunized with recombinant genetic vaccines (plasmid DNA and adenovirus) expressing this same T. cruzi antigen recognized, in addition to the immunodominant epitope, two other subdominant epitopes. This unexpected observation allowed us to test the protective role of the immune response to subdominant epitopes. This was accomplished by genetic vaccination of mice with mutated genes that did not express a functional immunodominant epitope. We found that these mice developed immune responses directed solely to the subdominant/cryptic CD8 T cell epitopes and a significant degree of protective immunity against infection mediated by CD8(+) T cells. We concluded that artificially broadening the T cell repertoire contributes to host resistance against infection, a finding that has implications for the host-parasite relationship and vaccine development.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Trypanosoma cruzi/microbiology , Animals , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Female , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/genetics , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Mice , Peptides/chemistry , Peptides/immunology , Tumor Necrosis Factor-alpha/metabolism
8.
Infect Immun ; 79(5): 2120-30, 2011 May.
Article in English | MEDLINE | ID: mdl-21357719

ABSTRACT

Recently, we described a heterologous prime-boost strategy using plasmid DNA followed by replication-defective human recombinant adenovirus type 5 as a powerful strategy to elicit long-lived CD8(+) T-cell-mediated protective immunity against experimental systemic infection of mice with a human intracellular protozoan parasite, Trypanosoma cruzi. In the present study, we further characterized the protective long-lived CD8(+) T cells. We compared several functional and phenotypic aspects of specific CD8(+) T cells present 14 or 98 days after the last immunizing dose and found the following: (i) the numbers of specific cells were similar, as determined by multimer staining or by determining the number of gamma interferon (IFN-γ)-secreting cells by enzyme-linked immunospot (ELISPOT) assay; (ii) these cells were equally cytotoxic in vivo; (iii) following in vitro stimulation, a slight decline in the frequency of multifunctional cells (CD107a(+) IFN-γ(+) or CD107a(+) IFN-γ(+) tumor necrosis factor alpha positive [TNF-α(+)]) was paralleled by a significant increase of CD107a singly positive cells after 98 days; (iv) the expression of several surface markers was identical, except for the reexpression of CD127 after 98 days; (v) the use of genetically deficient mice revealed a role for interleukin-12 (IL-12)/IL-23, but not IFN-γ, in the maintenance of these memory cells; and (vi) subsequent immunizations with an unrelated virus or a plasmid vaccine or the depletion of CD4(+) T cells did not significantly erode the number or function of these CD8(+) T cells during the 15-week period. From these results, we concluded that heterologous plasmid DNA prime-adenovirus boost vaccination generated a stable pool of functional protective long-lived CD8(+) T cells with an effector memory phenotype.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Chagas Disease/prevention & control , Immunologic Memory/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Vaccination/methods , Adenoviridae/genetics , Animals , Cell Separation , Female , Flow Cytometry , Genetic Vectors , Immunization, Secondary/methods , Mice , Mice, Inbred C57BL , Plasmids/immunology , Vaccines, DNA/immunology , Vaccines, Synthetic/immunology
9.
PLoS Pathog ; 6(11): e1001171, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-21079759

ABSTRACT

Influenza A virus causes annual epidemics which affect millions of people worldwide. A recent Influenza pandemic brought new awareness over the health impact of the disease. It is thought that a severe inflammatory response against the virus contributes to disease severity and death. Therefore, modulating the effects of inflammatory mediators may represent a new therapy against Influenza infection. Platelet activating factor (PAF) receptor (PAFR) deficient mice were used to evaluate the role of the gene in a model of experimental infection with Influenza A/WSN/33 H1N1 or a reassortant Influenza A H3N1 subtype. The following parameters were evaluated: lethality, cell recruitment to the airways, lung pathology, viral titers and cytokine levels in lungs. The PAFR antagonist PCA4248 was also used after the onset of flu symptoms. Absence or antagonism of PAFR caused significant protection against flu-associated lethality and lung injury. Protection was correlated with decreased neutrophil recruitment, lung edema, vascular permeability and injury. There was no increase of viral load and greater recruitment of NK1.1(+) cells. Antibody responses were similar in WT and PAFR-deficient mice and animals were protected from re-infection. Influenza infection induces the enzyme that synthesizes PAF, lyso-PAF acetyltransferase, an effect linked to activation of TLR7/8. Therefore, it is suggested that PAFR is a disease-associated gene and plays an important role in driving neutrophil influx and lung damage after infection of mice with two subtypes of Influenza A. Further studies should investigate whether targeting PAFR may be useful to reduce lung pathology associated with Influenza A virus infection in humans.


Subject(s)
Apoptosis , Influenza A Virus, H1N1 Subtype/pathogenicity , Lung Injury/metabolism , Lung Injury/virology , Orthomyxoviridae Infections/prevention & control , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Blotting, Western , Chickens , Dihydropyridines/pharmacology , Disease Models, Animal , Inflammation Mediators/metabolism , Influenza A Virus, H1N1 Subtype/genetics , Lung Injury/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Platelet Activating Factor/genetics , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Viral Load
10.
Vaccine ; 28(18): 3247-56, 2010 Apr 19.
Article in English | MEDLINE | ID: mdl-20189485

ABSTRACT

In this work, we explored an original vaccination protocol using recombinant influenza and adenovirus. We constructed recombinant influenza viruses harboring dicistronic NA segments containing the surface antigen 2 (SAG2) from Toxoplasma gondii under control of the duplicated 3' promoter. Recombinant influenza viruses were able to drive the expression of the foreign SAG2 sequence in cell culture and to replicate efficiently both in cell culture and in lungs of infected mice. In addition, mice primed with recombinant influenza virus and boosted with a recombinant adenovirus encoding SAG2 elicited both humoral and cellular immune responses specific for SAG2. Moreover, when immunized animals were challenged with the cystogenic P-Br strain of T. gondii, they displayed up to 85% of reduction in parasite burden. These results demonstrate the potential use of recombinant influenza vectors harboring the dicistronic segments in the development of vaccines against infectious diseases.


Subject(s)
Adenoviridae/genetics , Antigens, Protozoan/immunology , Immunization, Secondary/methods , Immunization/methods , Orthomyxoviridae/immunology , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Toxoplasma/immunology , Adjuvants, Immunologic/genetics , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Brain/parasitology , Cytokines/metabolism , Female , Genetic Vectors , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae/genetics , Orthomyxoviridae Infections/prevention & control , Protozoan Proteins/genetics , Protozoan Vaccines/genetics , Spleen/immunology , Survival Analysis , Toxoplasma/genetics , Toxoplasmosis/prevention & control
11.
Infect Immun ; 77(10): 4383-95, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19651871

ABSTRACT

A heterologous prime-boost strategy using plasmid DNA, followed by replication-defective recombinant adenovirus 5, is being proposed as a powerful way to elicit CD4(+) and CD8(+) T-cell-mediated protective immunity against intracellular pathogens. We confirmed this concept and furthered existing research by providing evidence that the heterologous prime-boost regimen using the gene encoding amastigote surface protein 2 elicited CD4(+) and CD8(+) T-cell-mediated protective immunity (reduction of acute parasitemia and prolonged survival) against experimental infection with Trypanosoma cruzi. Protective immunity correlated with the presence of in vivo antigen-specific cytotoxic activity prior to challenge. Based on this, our second goal was to determine the outcome of infection after heterologous prime-boost immunization of perforin-deficient mice. These mice were highly susceptible to infection. A detailed analysis of the cell-mediated immune responses in immunized perforin-deficient mice showed an impaired gamma interferon (IFN-gamma) secretion by immune spleen cells upon restimulation in vitro with soluble recombinant antigen. In spite of a normal numeric expansion, specific CD8(+) T cells presented several functional defects detected in vivo (cytotoxicity) and in vitro (simultaneous expression of CD107a/IFN-gamma or IFN-gamma/tumor necrosis factor alpha) paralleled by a decreased expression of CD44 and KLRG-1. Our final goal was to determine the importance of IFN-gamma in the presence of highly cytotoxic T cells. Vaccinated IFN-gamma-deficient mice developed highly cytotoxic cells but failed to develop any protective immunity. Our study thus demonstrated a role for perforin and IFN-gamma in a number of T-cell-mediated effector functions and in the antiparasitic immunity generated by a heterologous plasmid DNA prime-adenovirus boost vaccination strategy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chagas Disease/prevention & control , Interferon-gamma/immunology , Pore Forming Cytotoxic Proteins/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Animals , Female , Immunization, Secondary/methods , Interferon-gamma/deficiency , Mice , Mice, Inbred C57BL , Neuraminidase/genetics , Neuraminidase/immunology , Parasitemia/prevention & control , Pore Forming Cytotoxic Proteins/deficiency , Survival Analysis , Vaccination/methods , Vaccines, DNA/immunology , Vaccines, Synthetic/immunology
12.
Vaccine ; 27(41): 5644-53, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19635607

ABSTRACT

Immunisation with Amastigote Surface Protein 2 (asp-2) and trans-sialidase (ts) genes induces protective immunity in highly susceptible A/Sn mice, against infection with parasites of the Y strain of Trypanosoma cruzi. Based on immunological and biological strain variations in T. cruzi parasites, our goal was to validate our vaccination results using different parasite strains. Due to the importance of the CD8(+) T cells in protective immunity, we initially determined which strains expressed the immunodominant H-2K(k)-restricted epitope TEWETGQI. We tested eight strains, four of which elicited immune responses to this epitope (Y, G, Colombian and Colombia). We selected the Colombian and Colombia strains for our studies. A/Sn mice were immunised with different regimens using both T. cruzi genes (asp-2 and ts) simultaneously and subsequently challenged with blood trypomastigotes. Immune responses before the challenge were confirmed by the presence of specific antibodies and peptide-specific T cells. Genetic vaccination did not confer protective immunity against acute infection with a lethal dose of the Colombian strain. In contrast, we observed a drastic reduction in parasitemia and a significant increase in survival, following challenge with an otherwise lethal dose of the Colombia strain. In many surviving animals with late-stage chronic infection, we observed alterations in the heart's electrical conductivity, compared to naive mice. In summary, we concluded that immunity against T. cruzi antigens, similar to viruses and bacteria, may be strain-specific and have a negative impact on vaccine development.


Subject(s)
Chagas Disease/prevention & control , Glycoproteins/immunology , Neuraminidase/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/blood , Base Sequence , Cross Reactions , Epitopes, T-Lymphocyte/immunology , Female , Humans , Mice , Molecular Sequence Data , Parasitemia/prevention & control , Sequence Alignment , Survival Analysis , T-Lymphocytes/immunology
13.
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
14.
Hum Gene Ther ; 17(9): 898-908, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16972758

ABSTRACT

Protection against protozoan parasite Trypanosoma cruzi has been shown to be dependent on the induction of type 1 immune responses. Replication-deficient human type 5 recombinant adenoviruses have an unsurpassed ability to induce type 1 immune responses. Thus, we constructed two type 5 recombinant adenoviruses encoding parasite antigens trans-sialidase (rAdTS) and amastigote surface protein-2 (rAdASP2). Both antigens were genetically engineered to secrete recombinant products in order to induce both optimal antibody and T cell responses. Immunizations of mice with rAdASP2 and rAdTS induced high levels of serum antibodies specific for their recombinant products. In addition, both recombinant viruses were able to elicit a biased helper T cell type 1 (Th1) cellular immune response and a substantial CD8+ T cell-mediated immune response. Moreover, individual immunization with rAdASP2 or rAdTS induced high levels of protection against a challenge with live parasites. CD8+ T cells mediated, at least in part, such protection. Furthermore, when combined in the same inoculum, rAdTS plus rAdASP2 induced complete protection in all animals tested, even when challenges were performed 14 weeks after the last immunization. Taking together, these results show that recombinant adenoviruses expressing TS and ASP-2 antigens of T. cruzi are interesting candidates for the development of a vaccine against Chagas' disease.


Subject(s)
Adenoviridae/genetics , Glycoproteins/genetics , Neuraminidase/genetics , Recombination, Genetic , Trypanosoma cruzi/immunology , Trypanosomiasis/prevention & control , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred Strains , Mice, Knockout , Protozoan Vaccines/administration & dosage , Th1 Cells/immunology , Trypanosomiasis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...