Acessibilidade / Reportar erro

Swimming against the current: genetic vaccination against Trypanosoma cruzi infection in mice

Abstract

Vaccines have had an unquestionable impact on public health during the last century. The most likely reason for the success of vaccines is the robust protective properties of specific antibodies. However, antibodies exert a strong selective pressure and many microorganisms, such as the obligatory intracellular parasite Trypanosoma cruzi, have been selected to survive in their presence. Although the host develops a strong immune response to T. cruzi, they do not clear the infection and instead progress to the chronic phase of the disease. Parasite persistence during the chronic phase of infection is now considered the main factor contributing to the chronic symptoms of the disease. Based on this finding, containment of parasite growth and survival may be one method to avoid the immunopathology of the chronic phase. In this context, vaccinologists have looked over the past 20 years for other immune effector mechanisms that could eliminate these antibody-resistant pathogens. We and others have tested the hypothesis that non-antibody-mediated cellular immune responses (CD4+ Th1 and CD8+ Tc1 cells) to specific parasite antigens/genes expressed by T. cruzi could indeed be used for the purpose of vaccination. This hypothesis was confirmed in different mouse models, indicating a possible path for vaccine development.

Trypanosoma cruzi; vaccine; immunity


Swimming against the current: genetic vaccination against Trypanosoma cruzi infection in mice

Mauricio M RodriguesI,+ + Corresponding author: mrodrigues@unifesp.br ; Bruna C de AlencarI; Carla ClaserI; Fanny TzelepisI; Eduardo L SilveiraI; Filipe A HaollaI; Mariana R DominguezI; José Ronnie VasconcelosI, II, III

ICentro Interdisciplinar de Terapia Gênica (CINTERGEN)

IIDepartamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, Rua Mirassol 207, 04044-010 São Paulo, SP, Brasil

IIIDepartamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil

ABSTRACT

Vaccines have had an unquestionable impact on public health during the last century. The most likely reason for the success of vaccines is the robust protective properties of specific antibodies. However, antibodies exert a strong selective pressure and many microorganisms, such as the obligatory intracellular parasite Trypanosoma cruzi, have been selected to survive in their presence. Although the host develops a strong immune response to T. cruzi, they do not clear the infection and instead progress to the chronic phase of the disease. Parasite persistence during the chronic phase of infection is now considered the main factor contributing to the chronic symptoms of the disease. Based on this finding, containment of parasite growth and survival may be one method to avoid the immunopathology of the chronic phase. In this context, vaccinologists have looked over the past 20 years for other immune effector mechanisms that could eliminate these antibody-resistant pathogens. We and others have tested the hypothesis that non-antibody-mediated cellular immune responses (CD4+ Th1 and CD8+ Tc1 cells) to specific parasite antigens/genes expressed by T. cruzi could indeed be used for the purpose of vaccination. This hypothesis was confirmed in different mouse models, indicating a possible path for vaccine development.

Key words:Trypanosoma cruzi - vaccine - immunity

The population at risk of infection with obligatory intracellular parasites is estimated to be in the billions in individuals living in tropical and subtropical regions of the world where these parasites are endemic. Although the pattern of disease transmission, infection and mortality caused by these parasites has continued to change over the past 100 years, the global burden of disease is greater than it has ever been. In developing countries, the circumstances supporting disease transmission are unlikely to improve in the next 10 years. In light of these problems, it is essential to develop new strategies for the prevention and control of these diseases. Also, given that global warming will increase, we can anticipate that invertebrate hosts will occupy greater areas of the world and possibly increase transmission of insect-borne diseases.

Mass human vaccination against parasitic infection is currently not available. However, the first vaccine against human malaria is scheduled to become commercially available in 2010. Research in this field is considered to be important because in the future, vaccines may complement other strategies such as chemotherapy or vector control for the prevention and control of these parasitic diseases. As a result, immuno-parasitologists have been researching the mechanisms of host-protective immunity and its target antigens for the last 30 years.

In spite of the unquestionable impact of vaccination on public health during the last century, only two dozen vaccines are routinely used today. Mechanistically, we do not know how most vaccines provide pre-immunity. The most likely reason for the success of these vaccines is the robust protective properties of specific antibodies. In fact, the efficacy of commercial vaccines has always been measured as a function of the antibody immune response. Because antibodies are indeed so powerful, they exert a strong selective pressure on microorganisms. As expected, many microorganisms have been selected to survive in the presence of antibodies and ultimately use this selection to their advantage. Therefore, in many cases, the "robust" power of specific antibodies is not sufficient to eliminate the pathogen. In this context, over the last 25 years, vaccinologists have looked for other effector mechanisms that could either complement antibodies or act independently to eliminate these antibody-resistant pathogens.

The obligatory intracellular parasite Trypanosoma cruzi is responsible for the infection of millions of individuals across the Americas. Although hosts develop a strong immune response to T. cruzi, they do not clear the infection and instead progress to the chronic phase of the disease. Parasite persistence during the chronic phase of infection is now considered to be the main factor contributing to the chronic symptoms of the disease. Based on this finding, containment of parasite growth and survival may be the best route to avoiding chronic phase immunopathology. Prophylactic vaccination would help to reduce or completely eliminate the parasite burden, and thus represents a desirable method to restrict the development of chronic symptoms of the disease.

In the past, vaccination studies have failed because researchers have in most cases focused on inducing protective antibodies. Essentially, these studies confirmed that antibodies alone are unable to completely eliminate the parasites and that the antigens expressed in the axenic culture forms of the parasite are not the best targets for protective immunity. These failures have created a paradigm and led researchers to conclude that it would not be possible to generate an anti-T. cruzi vaccine. Over the past 10 years, we and others have challenged this tenet by testing the hypothesis that non-antibody-mediated cellular immune responses to the antigens expressed in the mammalian forms of the parasites could indeed be used for the purpose of vaccination. Using a mouse model of the disease, we confirmed this hypothesis by inducing protective immunity against T. cruzi infection specifically mediated by CD4+ Th1 and CD8+ Tc1 cells specific for antigens expressed by trypomastigotes and amastigotes of T. cruzi. While it is possible to obtain protective immunity against a challenge with a homologous parasite strain, the strain variation observed in certain immunodominant epitopes may impose a severe restriction on the development and use of this method for mass vaccination.

This review is not aimed at describing a product that may never exist; instead, our intention is to demonstrate how to elicit immunity to T. cruzi prior to infection in experimental models.

CD4+ and CD8+ T cells are important factors in mediating host survival during T. cruzi infection

The innate and adaptive immunity induced by T. cruzi is tailored to allow the infection while preventing the death of the host. After contact with the parasite, mammalian hosts initially undergo an acute phase of the infection, characterised by patient parasitemia. This period is critical for both the parasite and the host. The parasite has to concurrently establish a productive infection while at the same time keeping the host alive in order to maintain the parasite's life cycle. After the acute phase, the host's parasitemia levels decrease and the host progresses to the chronic phase of the infection. This latter phase is the period when most transmission to invertebrate hosts occurs. During the early stages of the acute phase, hosts survive because of strong innate immunity, mediated mostly by Toll-like receptors through the activation of the adaptor molecule MyD88 (Campos et al. 2004, Bafica et al. 2006, Bartholomeu et al. 2008). However, to avoid elimination, the parasite manages to significantly delay the onset of the CD8+ T cell-mediated immunity (CMI) response (Martin et al. 2006, Tzelepis et al. 2006, 2007). This manoeuvre is critical for the parasite to establish the infection. Only after the parasitemia reaches a critical point is a powerful CD8+ T CMI response triggered, drastically reducing the parasitemia and securing host survival. This CD8+ T CMI response, and possibly the CD4+ T cells and antibodies, collectively push the mouse into the long chronic phase of infection, during which the parasite is capable of transmission to the invertebrate host.

The role of T-cell-mediated adaptive immunity in the naturally acquired response to T. cruzi infection was determined by the use of genetically modified mice that express neither major histocompatibility complex (MHC) class I and II nor CD4 and CD8 molecules. These genetically deficient mouse strains are highly susceptible to acute infection because they are unable to control parasitemia (Rodrigues et al. 2003, Tarleton 2007). While MHC I- and CD8- deficient mice lack only CD8+ T CMI responses, MHC class II or CD4 deficiency leads to a more general failure, with reductions in both the antibody and the CD8 immune responses (Rottenberg et al. 1993, Tarleton 2007, Tzelepis et al. 2007). Therefore, CD4+ T cells may play an extremely important role in generating different mechanisms of protection. However, their function as an effector mechanism during naturally acquired immunity is less clear, as CD8-deficient mice are highly susceptible to infection with most parasite strains (Tarleton 2007, Tzelepis et al. 2007).

During human infection, T cells specific for T. cruzi can be detected in most individuals who are in the chronic phase of infection. Although it is difficult to definitively prove that these cells are important to host resistance, it is plausible that they exert an anti-parasitic effect similar to that seen in the mouse model of infection. During the chronic phase of the disease, most individuals have strong CMI to parasite antigens (Morato et al. 1986, Cetron et al. 1993, Dutra et al. 1996, Barros-Mazon et al. 1997, Wizel et al. 1998b, Ribeirão et al. 2000, Fonseca et al. 2005, Albareda et al. 2006, Alvarez et al. 2008). Events causing depression of CMI, such as immuno-suppressive treatment or HIV infection, lead to the relatively fast development of symptomatic forms of the disease in a percentage of chagasic patients (Rocha et al. 1994). In some cases, the reappearance of patient parasitemia characteristic of the acute phase of infection is observed (Sartori et al. 2002). This evidence suggests that CMI may help to maintain the low levels of parasites observed in chronic chagasic patients.

Characterization of the in vivo effector mechanisms mediated by protective T cells

Although there is evidence showing that T cells participate in the protective immunity against experimental T. cruzi infection, the precise mechanism(s) used by these cells to eliminate T. cruzi has(ve) not been defined. The fact that T cells secrete IFN-γ suggests that this mechanism may play a role in eliminating intracellular forms of the parasite. IFN-γ is an important mediator of naturally-acquired immunity against the infection. Genetically deficient mice that do not express the IFN-γ receptor or Stat4 are unable to control T. cruzi infection (Holscher et al.1998, Tarleton et al. 2000). However, no direct link between IFN-γ secretion by T cells and the in vivo anti-parasitic activity of these cells has been established to date.

The IFN-γ-dependent mechanisms are in part mediated by the production of nitric oxide (NO). In the absence of IFN-γ receptors, NO is not produced by macrophages, suggesting that IFN-γ is the main inducer of NO synthase 2 (NOS2) activation. The evidence that NO is critical for eliminating T. cruzi during acute experimental infection was obtained with genetically deficient mice that do not express NOS2. These animals are highly susceptible to infection with different parasite strains (Holscher et al. 1998, Rodrigues et al. 2000).

In addition to producing IFN-γ, CD8+ T cells can lyse host cells infected with T. cruzi, or secrete other potentially active mediators such as TNF-αa, granulisin or a number of different chemokines (Munoz-Fernandez et al. 1992, Aliberti et al. 1999, Stenger et al. 1999). In fact, CD8+ T cells specific for amastigote or trypomastigote antigens are capable of lysing non-phagocytic cells infected with T. cruzi in vitro (Wizel et al. 1998b, Low et al. 1998). However, it is unclear whether lysis and DNA degradation of infected target cells by CD8+ T cells can be effective in restricting T. cruzi infection in vivo. Genetically deficient mice that do not make perforin or granzyme B are not more susceptible to infection than wild-type mice after infection with the Brazil strain of T. cruzi (Kumar & Tarleton 1998). However, other T. cruzi strains behave differently and perforin-deficient mice are highly susceptible to infection (Tzelepis et al. 2006). Based on these observations, perforin-dependent and independent mechanism(s) provide resistance against T. cruzi infection in mice. The elucidation of the anti-parasitic mechanisms mediated by T. cruzi-specific CD8+ T cells will require further investigation using more complex experimental models.

Target antigens of the protective immune response and design of recombinant subunit vaccines for experimental trials

T. cruzi antigens recognised by immune sera from immune or infected humans or animals were the first antigens to be described at the molecular level. This characterisation allowed researchers to conduct studies using recombinant proteins based on isolated, antigen-specific genes. These isolated genes included members of the large trans-sialidase (TS) surface protein family expressed mainly in the infective trypomastigote and amastigote forms of the parasite. The second group of genes belonged to the family of cysteine-proteases (cruzipain) expressed in all of the different forms of the parasite. Other antigens formed a heterogeneous group including molecules such as the flagellar calcium-binding protein, paraflagellar rod protein-2, LYT-1 antigen, Ribosomal Protein L7a-Like Protein and KMP11, among others.

Several delivery antigens were used successfully to induce T. cruzi-specific T lymphocytes and protective immunity against an experimental infection. They included recombinant proteins mixed in the presence of distinct adjuvants, plasmid DNA and, more recently, recombinant viruses and bacteria. Table displays a list matching the different antigens with their delivery system.

As shown in Table, protective immune response in the mouse model as measured by a reduction in acute phase parasitemia, tissue parasitism and mortality can be elicited by a variety of antigens using distinct delivery systems. In most cases, immunity elicited by these antigens is associated with type I immune response, generated by IFN-γ producing CD4+ and/or CD8+ T cells. In a few cases, some of the mechanisms of induction of protective immunity were investigated. Following intranasal immunisation with (TS) in the presence of the TLR-9 activator CpG ODN, B cells are critical to inducing type I immune response mediated by CD4+ and CD8+ T cells (Hoft et al. 2007). In this same model, the absence of CD4+ or CD8+ T cells also renders the vaccinated animals completely susceptible to infection. Because these animals were genetically deficient, these cells can be required for the induction or the effector phase of the immune response, or both. Similarly, CD8- deficient mice failed to generate protection after immunisation with native Par-2 protein emulsified in CFA or recombinant adenovirus expressing TS or ASP-2 genes (Wrightsman et al. 2002, Machado et al. 2006).

The effector mechanisms responsible for the protective immunity have been evaluated in a few experimental models. Upon plasmid immunisation, depletion of either CD4+ or CD8+ T cells completely reversed protective immunity, thus demonstrating a non-overlapping role for these two subpopulations (Katae et al. 2002, Vasconcelos et al. 2004). Following vaccination with recombinant protein of ASP-2 in alum and CpG ODN, only depletion of CD8+, but not CD4+, T cells reversed protective immunity (Araújo et al. 2005). Finally, by using a single T. cruzi epitope recognised by CD8+ T cells, Miyahira et al. (2005) elicited a protective immune response using a heterologous prime-boost strategy with recombinant adenovirus and vaccinia virus. Although only a few experimental systems found type 1 CD4+ and CD8+ T cells to be necessary, these findings confirm the general paradigm that CD4+ and CD8+ T cells do play a key role in CMI. The question as to whether other cells types are also critical for the adaptive immunity induced by these recombinant vaccines requires additional investigation. In addition, it will be important to systematically define the phenotype and function of the specific T cells in order to establish the immunological status of vaccinated individuals prior to their encounter with the pathogen.

In spite of clear evidence that immunisation with T. cruzi antigens can provide protective immunity as measured by a reduction in acute phase parasitemia, tissue parasitism and mortality, it is not clear whether immunisation will lead to either remission or a cure of the chronic phase symptoms of the disease. To determine the role of immunisation in reducing chronic phase disease symptoms, a number of experiments using different animal models must be performed. In many of the models described above, tissue inflammation and parasitism in the late chronic phase were significantly reduced following prophylactic vaccination (Garg & Tarleton 2002, Vasconcelos et al. 2004, Araújo et al. 2005). Therefore it is possible that prophylactic vaccinations indeed halt the development of the chronic phase immunopathologies. Nevertheless, the most compelling evidence of a vaccine's ability to reduce the immunopathology was obtained by therapeutic immunisation with T. cruzi genes encoding the TSA and Tc24 genes (Dunonteil et al. 2004). Whether these results are reproducible using different combinations of mouse and parasite strains remains to be seen.

Evasion of T-cell immune responses

Viruses are the best known class of pathogens that use a variety of mechanisms to evade the immune recognition by MHC class I restricted CD8+ T cells. Some of these strategies may function at the level of antigen presentation by the infected cells such as: (i) downregulation of MHC class I expression, (ii) interference with cytosolic proteolysis, (iii) inhibition of peptide transport to the endoplasmic reticulum (ER), (iv) retention and destruction of MHC class I molecules in the ER and (v) presentation of mutant epitopes that do not bind to MHC I class molecules. Under these circumstances, infected cells have a limited amount of peptide/MHC-class I complexes on their surface, making it difficult for them to be recognised by specific CD8+ T cells. A detailed review and examples of these evasion mechanisms can be found in Antoniou and Powis (2008).

Alternatively, escape mechanisms can interfere with the recognition by MHC class I restricted CD8+ T cells. For example, mutant viruses can be selected with amino acid variations that retain binding to MHC class I molecules but result in reduced recognition by T cells or generate antagonistic peptides that inhibit activation of specific T cells by the MHC-peptide complex (Ploegh 1998, Antoniou & Powis 2008).

Most recently, the literature has described viral proteins that interfere with the effector mechanisms mediated by T cells. For example, poxviruses encode several soluble cytokine receptors that bind to and block the activity of IFN-α, β and γ, TNF-α, and IL-1. Also, poxviruses express a protein termed crmA that functionally inhibits Asp-specific cysteine proteases (caspases), therefore blocking apoptosis induced by CTL, TNF or Fas (Ploegh 1998). It is not known whether T. cruzi parasites have evolved to employ these immune evasion mechanisms. To date, not a single evasion mechanism has been confirmed in terms of a CD8+ T CMI response to T. cruzi. This lack of information most likely reflects the fact that the CD8+ T CMI response has not been thoroughly investigated.

Recently, the identification of CD8+ T cell epitopes derived against proteins of the TS superfamily allowed the comparison of the immunodominant responses resulting from experimental infection in inbred mice following challenge with distinct parasite strains (Martin et al. 2006, Tzelepis et al. 2006, 2007, 2008). CD8+ T cells of a single mouse strain (C57BL/6 mice) infected with different parasite strains recognised distinct peptides as the immunodominant epitope. The diversity of the immunodominant response between strains presents a significant challenge for developing vaccines. A vaccine would require a number of CD8+ epitopes that cover not only the different human MHC haplotypes but also the different parasite strains. Alternatively, the use of sub-dominant epitopes could favour the protective immune responses without requiring so many epitopes (Rodrigues et al. 2009).

Finally, the presence of a strong CD8+ T cell immune response may select variants that no longer act as a target for these protective CD8+ T cells as is the case with HCV or HIV (Tester et al. 2005, Watkins 2008). This hypothesis was tested once following vaccination with TS in the presence of CpG ODN. Parasites isolated from immune mice did not show any increase in resistance to anti-TS immune responses, indicating that antigenically distinct forms of the parasites were not selected by the strong protective immune response (Hoft et al. 2007). The question as to whether the absence of selectively imposed variations has an effect on CD8+ T cell immune evasion should be evaluated in more experimental models.

Perspectives

The knowledge that T cells are essential for efficient immunity to T. cruzi has raised important questions with basic and applied implications. Several gaps in our basic knowledge are expected to be filled in the years to come. Some of the most relevant questions are: (i) how are specific CD8+ T cells induced and maintained in vivo? (ii) What are the main effector mechanisms operating in vivo against the different forms of the parasite? (iii) Are there specific or general mechanisms of immune evasion used by each strain? These questions are even more relevant and much more complex and difficult to study in humans due to the obvious technical limitations.

From the applied point of view, it is now clear that a number of different delivery systems can be efficient in terms of the expansion of protective T lymphocytes. Vaccination trials using non-human primates aimed at inducing T CMI have yet to be conducted and should provide important information about the feasibility of this strategy to develop the first vaccines against T. cruzi infection.

It is possible that these studies on the immune response elicited following vaccination with recombinant proteins may contribute to the development of new strategies for the prevention or treatment of T. cruzi infection. However, due to the complex nature of this parasitic disease, we believe that it is too early to anticipate the utility of this strategy in humans.

Received 10 March 2009

Accepted 30 April 2009

Financial support: FAPESP, INTEV-CNPq (MMR), CNPq (MMR, ELS), FAPESP (BCGA, JRV, CC, FT, FAH, MDR)

  • Albareda MC, Laucella SA, Alvarez MG, Armenti AH, Bertochi G, Tarleton RL, Postan M 2006. Trypanosoma cruzi modulates the profile of memory CD8+ T cells in chronic Chagas' disease patients. Int Immunol 18: 465-471.
  • Aliberti JC, Machado FS, Souto JT, Campanelli AP, Teixeira MM, Gazzinelli RT, Silva JS 1999. beta-Chemokines enhance parasite uptake and promote nitric oxide-dependent microbiostatic activity in murine inflammatory macrophages infected with Trypanosoma cruzi Infect Immun 67: 4819-4826.
  • Alvarez MG, Postan M, Weatherly DB, Albareda MC, Sidney J, Sette A, Olivera C, Armenti AH, Tarleton RL, Laucella SA 2008. HLA class I-T cell epitopes from trans-sialidase proteins reveal functionally distinct subsets of CD8 T cells in chronic Chagas disease. PLoS Negl Trop Dis 2: e288.
  • Antoniou AN, Powis SJ 2008. Pathogen evasion strategies for the major histocompatibility complex class I assembly pathway. Immunology 124: 1-12.
  • Araújo AF, de Alencar BC, Vasconcelos JR, Hiyane MI, Marinho CR, Penido ML, Boscardin SB, Hoft DF, Gazzinelli RT, Rodrigues MM 2005. CD8+-T-cell-dependent control of Trypanosoma cruzi infection in a highly susceptible mouse strain after immunization with recombinant proteins based on amastigote surface protein 2. Infect Immun 73: 6017-6025.
  • Bafica A, Santiago HC, Goldszmid R, Ropert C, Gazzinelli RT, Sher A 2006. Cutting edge: TLR9 and TLR2 signaling together account for MyD88-dependent control of parasitemia in Trypanosoma cruzi infection. J Immunol 177: 3515-3519.
  • Barros-Mazon S, Guariento ME, Abrahamsohn IA 1997. IL-12 enhances proliferation of peripheral blood mononuclear cells from Chagas' disease patients to Trypanosoma cruzi antigen. Immunol Lett 57: 39-45.
  • Bartholomeu DC, Ropert C, Melo MB, Parroche P, Junqueira CF, Teixeira SM, Sirois C, Kasperkovitz P, Knetter CF, Lien E, Latz E, Golenbock DT, Gazzinelli RT 2008. Recruitment and endo-lysosomal activation of TLR9 in dendritic cells infected with Trypanosoma cruzi J Immunol 181: 1333-1344.
  • Bhatia V, Garg NJ 2008. Previously unrecognized vaccine candidates control Trypanosoma cruzi infection and immunopathology in mice. Clin Vaccine Immunol 15: 1158-1164.
  • Boscardin SB, Kinoshita SS, Fujimura AE, Rodrigues MM 2003. Immunization with cDNA expressed by amastigotes of Trypanosoma cruzi elicits protective immune response against experimental infection. Infect Immun 71: 2744-2757.
  • Campos MA, Closel M, Valente EP, Cardoso JE, Akira S, Alvarez-Leite JI, Ropert C, Gazzinelli RT 2004. Impaired production of proinflammatory cytokines and host resistance to acute infection with Trypanosoma cruzi in mice lacking functional myeloid differentiation factor 88. J Immunol 172: 1711-1718.
  • Cazorla SI, Becker PD, Frank FM, Ebensen T, Sartori MJ, Corral RS, Malchiodi EL, Guzmán CA 2008. Oral vaccination with Salmonella enterica as a cruzipain-DNA delivery system confers protective immunity against Trypanosoma cruzi Infect Immun 76: 324-333.
  • Cazorla SI, Frank FM, Becker PD, Corral RS, Guzmán CA, Malchiodi EL 2008. Prime-boost immunization with cruzipain co-administered with MALP-2 triggers a protective immune response able to decrease parasite burden and tissue injury in an experimental Trypanosoma cruzi infection model. Vaccine 26: 1999-2009.
  • Cetron MS, Basilio FP, Moraes AP, Sousa AQ, Paes JN, Kahn SJ, Wener MH, Van Voorhis WC 1993. Humoral and cellular immune response of adults from Northeastern Brazil with chronic Trypanosoma cruzi infection: depressed cellular immune response to T. cruzi antigen among Chagas' disease patients with symptomatic versus indeterminate infection. Am J Trop Med Hyg 49: 370-382.
  • Claser C, Espíndola NM, Sasso G, Vaz AJ, Boscardin SB, Rodrigues MM 2007. Immunologically relevant strain polymorphism in the Amastigote Surface Protein 2 of Trypanosoma cruzi Microbes Infect 9: 1011-1019.
  • Costa F, Franchin G, Pereira-Chioccola VL, Ribeirão M, Schenkman S, Rodrigues MM 1998. Immunization with a plasmid DNA containing the gene of trans-sialidase reduces Trypanosoma cruzi infection in mice. Vaccine 16: 768-774.
  • de Alencar BC, Araújo AF, Penido ML, Gazzinelli RT, Rodrigues MM 2007. Cross-priming of long lived protective CD8+ T cells against Trypanosoma cruzi infection: importance of a TLR9 agonist and CD4+ T cells. Vaccine 25: 6018-6027.
  • Dumonteil E, Escobedo-Ortegon J, Reyes-Rodriguez N, Arjona-Torres A, Ramirez-Sierra MJ 2004. Immunotherapy of Trypanosoma cruzi infection with DNA vaccines in mice. Infect Immun 72: 46-53.
  • Dutra WO, da Luz ZM, Cançado JR, Pereira ME, Brigido-Nunes RM, Galvão LM, Colley DG, Brener Z, Gazzinelli G, Carvalho-Parra JF 1996. Influence of parasite presence on the immunologic profile of peripheral blood mononuclear cells from chagasic patients after specific drug therapy. Parasite Immunol 18: 579-585.
  • Fonseca SG, Moins-Teisserenc H, Clave E, Ianni B, Nunes VL, Mady C, Iwai LK, Sette A, Sidney J, Marin ML, Goldberg AC, Guilherme L, Charron D, Toubert A, Kalil J, Cunha-Neto E 2005. Identification of multiple HLA-A*0201-restricted cruzipain and FL-160 CD8+ epitopes recognized by T cells from chronically Trypanosoma cruzi-infected patients. Microbes Infect 7: 688-697.
  • Fontanella GH, De Vusser K, Laroy W, Daurelio L, Nocito AL, Revelli S, Contreras R 2008. Immunization with an engineered mutant trans-sialidase highly protects mice from experimental Trypanosoma cruzi infection: a vaccine candidate. Vaccine 26: 2322-2234.
  • Fralish BH, Tarleton RL 2003. Genetic immunization with LYT1 or a pool of trans-sialidase genes protects mice from lethal Trypanosoma cruzi infection. Vaccine 21: 3070-3080.
  • Frank FM, Petray PB, Cazorla SI, Muñoz MC, Corral RS, Malchiodi EL 2003. Use of a purified Trypanosoma cruzi antigen and CpG oligodeoxynucleotides for immunoprotection against a lethal challenge with trypomastigotes. Vaccine 22: 77-86.
  • Fujimura AE, Kinoshita SS, Pereira-Chioccola VL, Rodrigues MM 2001. DNA sequences encoding CD4+ and CD8+ T-cell epitopes are important for efficient protective immunity induced by DNA vaccination with a Trypanosoma cruzi gene. Infect Immun 69: 5477-5486.
  • Garg N, Tarleton RL 2002. Genetic immunization elicits antigen-specific protective immune responses and decreases disease severity in Trypanosoma cruzi infection. Infect Immun 70: 5547-5555.
  • Hoft DF, Eickhoff CS, Giddings OK, Vasconcelos JR, Rodrigues MM 2007. Trans-sialidase recombinant protein mixed with CpG motif-containing oligodeoxynucleotide induces protective mucosal and systemic Trypanosoma cruzi immunity involving CD8+ CTL and B cell-mediated cross-priming. J Immunol 179: 6889-6900.
  • Holscher C, Kohler G, Muller U, Mossmann H, Schaub GA, Brombacher F 1998. Defective nitric oxide effector functions lead to extreme susceptibility of Trypanosoma cruzi-infected mice deficient in gamma interferon receptor or inducible nitric oxide synthase. Infect Immun 66: 1208-1215.
  • Katae M, Miyahira Y, Takeda K, Matsuda H, Yagita H, Okumura K, Takeuchi T, Kamiyama T, Ohwada A, Fukuchi Y, Aoki T 2002. Coadministration of an interleukin-12 gene and a Trypanosoma cruzi gene improves vaccine efficacy Infect Immun 70: 4833-4840.
  • Kumar S, Tarleton RL 1998. The relative contribution of antibody production and CD8+ T cell function to immune control of Trypanosoma cruzi Parasite Immunol 20: 207-216.
  • Low HP, Santos MA, Wizel B, Tarleton RL 1998. Amastigote surface proteins of Trypanosoma cruzi are targets for CD8+ CTL. J Immunol 160: 1817-1823.
  • Machado AV, Cardoso JE, Claser C, Rodrigues MM, Gazzinelli RT, Bruna-Romero O 2006. Long-term protective immunity induced against Trypanosoma cruzi infection after vaccination with recombinant adenoviruses encoding amastigote surface protein-2 and trans-sialidase. Hum Gene Ther 17: 898-908.
  • Martin DL, Weatherly DB, Laucella SA, Cabinian MA, Crim MT, Sullivan S, Heiges M, Craven SH, Rosenberg CS, Collins MH, Sette A, Postan M, Tarleton RL 2006. CD8+ T-Cell responses to Trypanosoma cruzi are highly focused on strain-variant trans-sialidase epitopes. PLoS Pathog 2: e77.
  • Miyahira Y, Akiba H, Katae M, Kubota K, Kobayashi S, Takeuchi T, García-Sastre A, Fukuchi Y, Okumura K, Yagita H, Aoki T 2003. Cutting edge: a potent adjuvant effect of ligand to receptor activator of NF-kappa B gene for inducing antigen-specific CD8+ T cell response by DNA and viral vector vaccination. J Immunol 171: 6344-6348.
  • Miyahira Y, Takashima Y, Kobayashi S, Matsumoto Y, Takeuchi T, Ohyanagi-Hara M, Yoshida A, Ohwada A, Akiba H, Yagita H, Okumura K, Ogawa H 2005. Immune responses against a single CD8+-T-cell epitope induced by virus vector vaccination can successfully control Trypanosoma cruzi infection. Infect Immun 73: 7356-7365.
  • Morato MJ, Brener Z, Cançado JR, Nunes RM, Chiari E, Gazzinelli G 1986. Cellular immune responses of chagasic patients to antigens derived from different Trypanosoma cruzi strains and clones. Am J Trop Med Hyg 35: 505-511.
  • Munoz-Fernandez MA, Fernandez MA, Fresno M 1992. Synergism between tumor necrosis factor-alpha and interferon-gamma on macrophage activation for the killing of intracellular Trypanosoma cruzi through a nitric oxide-dependent mechanism. Eur J Immunol 22: 301-307.
  • Planelles L, Thomas MC, Alonso C, López MC 2001. DNA immunization with Trypanosoma cruzi HSP70 fused to the KMP11 protein elicits a cytotoxic and humoral immune response against the antigen and leads to protection. Infect Immun 69: 6558-6563.
  • Ploegh HL 1998. Viral strategies of immune evasion. Science 280: 248-253.
  • Ribeirão M, Pereira-Chioccola VL, Rénia L, Augusto Fragata Filho A, Schenkman S, Rodrigues MM 2000. Chagasic patients develop a type 1 immune response to Trypanosoma cruzi trans-sialidase. Parasite Immunol; 22: 49-53
  • Rocha A, de Meneses AC, da Silva AM, Ferreira MS, Nishioka SA, Burgarelli MK, Almeida E, Turcato Júnior G, Metze K, Lopes ER 1994. Pathology of patients with Chagas' disease and acquired immunodeficiency syndrome. Am J Trop Med Hyg 50: 261-268.
  • Rodrigues MM, Boscardin SB, Vasconcelos JR, Hiyane MI, Salay G, Soares I 2003. Importance of CD8 T cell-mediated immune response during intracellular parasitic infections and its implications for the development of effective vaccines. An Acad Bras Cienc 75: 443-468.
  • Rodrigues MM, de Alencar BC, Claser C, Tzelepis F 2009. Immunodominance: a new hypothesis to explain parasite escape and host/parasite equilibrium leading to the chronic phase of Chagas' disease? Braz J Med Biol Res 42: 220-223.
  • Rodrigues MM, Riberão M, Boscardin SB 2000. CD4 Th1 but not Th2 clones efficiently activate macrophages to eliminate Trypanosoma cruzi through a nitric oxide dependent mechanism. Immunol Lett 73: 43-50.
  • Rottenberg ME, Bakhiet M, Olsson T, Kristensson K, Mak T, Wigzell H, Orn A 1993. Differential susceptibilities of mice genomically deleted of CD4 and CD8 to infections with Trypanosoma cruzi or Trypanosoma brucei Infect Immun 61: 5129-5133.
  • Sartori AM, Neto JE, Nunes EV, Braz LM, Caiaffa-Filho HH, Oliveira Oda C Jr, Neto VA, Shikanai-Yasuda MA 2002. Trypanosoma cruzi parasitemia in chronic Chagas disease: comparison between human immunodeficiency virus (HIV)-positive and HIV-negative patients. J Infect Dis 186: 872-875.
  • Schnapp AR, Eickhoff CS, Sizemore D, Curtiss R 3rd, Hoft DF 2002. Cruzipain induces both mucosal and systemic protection against Trypanosoma cruzi in mice. Infect Immun 70: 5065-5074.
  • Sepulveda P, Hontebeyrie M, Liegeard P, Mascilli A, Norris KA 2000. DNA-based immunization with Trypanosoma cruzi complement regulatory protein elicits complement lytic antibodies and confers protection against Trypanosoma cruzi infection. Infect Immun 68: 4986-4991.
  • Silveira EL, Claser C, Haolla FA, Zanella LG, Rodrigues MM 2008. Novel protective antigens expressed by Trypanosoma cruzi amastigotes provide immunity to mice highly susceptible to Chagas' disease. Clin Vaccine Immunol 15: 1292-1300.
  • Stenger S, RosaT JP, Bloom BR, Krensky AM, Modlin RL 1999. Granulysin: a lethal weapon of cytolytic T cells. Immunol Today 20: 390-394.
  • Tarleton RL 2007. Immune system recognition of Trypanosoma cruzi Curr Opin Immunol 19: 430-434.
  • Tarleton RL, Grusby MJ, Zhang L 2000. Increased susceptibility of Stat4-deficient and enhanced resistance in Stat6-deficient mice to infection with Trypanosoma cruzi J Immunol 165: 1520-1525.
  • Tester I, Smyk-Pearson S, Wang P, Wertheimer A, Yao E, Lewinsohn DM, Tavis JE, Rosen HR 2005. Immune evasion versus recovery after acute hepatitis C virus infection from a shared source. J Exp Med 201: 1725-1731.
  • Tzelepis F, de Alencar BC, Penido ML, Claser C, Machado AV, Bruna-Romero O, Gazzinelli RT, Rodrigues MM 2008. Infection with Trypanosoma cruzi restricts the repertoire of parasite-specific CD8+ T cells leading to immunodominance. J Immunol 180: 1737-1748.
  • Tzelepis F, de Alencar BC, Penido ML, Gazzinelli RT, Persechini PM, Rodrigues MM 2006. Distinct kinetics of effector CD8+ cytotoxic T cells after infection with Trypanosoma cruzi in naive or vaccinated mice. Infect Immun 74: 2477-2481.
  • Tzelepis F, Persechini PM, Rodrigues MM 2007. Modulation of CD4+ T cell-dependent specific cytotoxic CD8+ T cells differentiation and proliferation by the timing of increase in the pathogen load. PLoS ONE 25 2: e393.
  • Vasconcelos JR, Hiyane MI, Marinho CR, Claser C, Machado AM, Gazzinelli RT, Bruña-Romero O, Alvarez JM, Boscardin SB, Rodrigues MM 2004. Protective immunity against Trypanosoma cruzi infection in a highly susceptible mouse strain after vaccination with genes encoding the amastigote surface protein-2 and trans-sialidase. Hum Gene Ther 15: 878-886.
  • Watkins DI 2008. The hope for an HIV vaccine based on induction of CD8+ T lymphocytes - A review. Mem Inst Oswaldo Cruz 103: 119-129.
  • Wizel B, Nunes M, Tarleton RL 1997. Identification of Trypanosoma cruzi trans-sialidase family members as targets of protective CD8+ TC1 responses. J Immunol 159:6120-6130.
  • Wizel B, Garg N, Tarleton RL 1998a. Vaccination with trypomastigote surface antigen 1-encoding plasmid DNA confers protection against lethal Trypanosoma cruzi infection. Infect Immun 66: 5073-5081.
  • Wizel B, Palmieri M, Mendonza C, Arana B, Sidney J, Sette A, Tarleton R 1998b. Human infection with Trypanosoma cruzi induces parasite antigen-specific cytotoxic T lymphocyte responses. J Clin Invest 102: 1062-1071.
  • Wrightsman RA, Luhrs KA, Fouts D, Manning JE 2002. Paraflagellar rod protein-specific CD8+ cytotoxic T lymphocytes target Trypanosoma cruzi-infected host cells. Parasite Immunol 24: 401-412.
  • Wrightsman RA, Manning JE 2000. Paraflagellar rod proteins administered with alum and IL-12 or recombinant adenovirus expressing IL-12 generates antigen-specific responses and protective immunity in mice against Trypanosoma cruzi Vaccine 18: 1419-1427.
  • Zapata-Estrella H, Hummel-Newell C, Sanchez-Burgos G, Escobedo-OrtegonJ, Ramirez-Sierra MJ, Arjona-Torres A, Dumonteil E 2006. Control of Trypanosoma cruzi infection and changes in T-cell populations induced by a therapeutic DNA vaccine in mice. Immunol Lett 103: 186-191.
  • +
    Corresponding author:
  • Publication Dates

    • Publication in this collection
      21 July 2009
    • Date of issue
      July 2009

    History

    • Accepted
      30 Apr 2009
    • Received
      10 Mar 2009
    Instituto Oswaldo Cruz, Ministério da Saúde Av. Brasil, 4365 - Pavilhão Mourisco, Manguinhos, 21040-900 Rio de Janeiro RJ Brazil, Tel.: (55 21) 2562-1222, Fax: (55 21) 2562 1220 - Rio de Janeiro - RJ - Brazil
    E-mail: memorias@fiocruz.br