点击显示 收起
Departments of Biochemistry and Immunology and Pathology, School of Medicine of Ribeiro Preto, University of So Paulo, Ribeiro Preto, So Paulo
Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
The pathogenesis of myocarditis during Trypanosoma cruzi infection is poorly understood. We investigated the role played by chemokine receptor 5 (CCR5) in the influx of T cells to the cardiac tissue of T. cruziinfected mice. mRNA and protein for the CCR5 ligands CCL3, CCL4, and CCL5 were detected in the hearts of infected mice in association with CD4+ and CD8+ T cells. There was a high level of CCR5 expression on CD8+ T cells in the hearts of infected mice. Moreover, CCR5 expression on CD8+ T cells was positively modulated by T. cruzi infection. CCR5-deficient mice infected with T. cruzi experienced a dramatically inhibited migration of T cells to the heart and were also more susceptible to infection. These results suggest that CCR5 and its ligands play a central role in the control of T cell influx in T. cruziinfected mice. Knowledge of the mechanisms that trigger and control the migration of cells to the heart in patients with Chagas disease may help in the design of drugs that prevent myocarditis and protect against the development of severe disease.
Trypanosoma cruzi is an obligatory intracellular protozoan parasite that causes Chagas disease, an important public-health problem in Latin America. In studies of infection, there is significant blood parasitemia, tissue parasitism, and tissue inflammation and a great degree of lethality, all of which are dependent on the strain of the parasite, the dose of the inoculum, and the species being studied. Control of the infection is eventually achieved. Several studies have reported an essential role for CD4+ [13] and CD8+ [3, 4] T cells and for interferon (IFN) and tumor necrosis factor (TNF) [58] in the control of acute infection. However, it is still not known how these types of cells migrate to sites of tissue infection and interact with inflammatory mediators at these sites to mediate the control of parasite replication.
We recently reported that T. cruziinfected macrophages and cardiomyocytes expressed mRNA for a range of chemokines, including CCL3 (macrophage inflammatory protein 1), CCL4 (MIP-1), and CCL5 (RANTES) [9, 10]. In these studies, not only were these chemokines expressed, but T. cruziinfected macrophages and cardiomyocytes also expressed NO, to mediate NO-dependent killing in vitro [913]. Moreover, we showed that mRNA for these chemokines was also expressed during acute infection in mice and that cytokines (i.e., TNF- and IFN-) known to participate in the control of infection modulated the expression of chemokines [1416]. Given that chemokines are well known for their ability to induce the migration of leukocytes and the activation of immune cells, these previous studies suggested that, in addition to facilitating the production of NO, the expression of chemokines could drive the migration of inflammatory and immune cells to sites of parasite infection.
The biological activities of chemokines are mediated by their interactions with cell-surface receptors that belong to the structurally related 7-transmembrane-domain superfamily of proteins [17]. CCR5 mediates the activity of the chemokines CCL3, CCL4, and CCL5, whose mRNA is found in vitro after infection of macrophages and cardiomyocytes with T. cruzi [9, 10] and in vivo after infection of macrophages and cardiomyocytes with the Colombiana strain [15]. As such, these chemokines may have a relevant role in driving the selective migration of CCR5+ immune cells toward affected tissues. Here, we confirm the expression of CCL3, CCL4, and CCL5 in mice infected with the Y strain of T. cruzi. Moreover, using CCR5-deficient (CCR5-/-) mice, we evaluate the role played by the interaction between CCR5 and its ligands in protection against T. cruzi infection and in the development and maintenance of myocarditis.
MATERIALS AND METHODS
Mice.
A breeding pair of mice with disruption of the CCR5 gene [18] was obtained from Jackson Laboratories. Breeding stock backcrossed on C57BL/6 was obtained, and the genotype of CCR5 in mice was determined by polymerase chain reaction (PCR) of DNA [19]. Female C57BL/6 wild-type (wt) and CCR5-/- mice, 68 weeks old, were bred and maintained in microisolator cages. All procedures were in accordance with international guidelines for the use of animals and received prior approval by the animal ethics committee of the School of Medicine of Ribeiro Preto.
Parasites and experimental infection.
The Y strain of T. cruzi was used in all experiments. For in vitro experiments, trypomastigote forms were grown and purified from a fibroblast cell line (LLC-MK2). In all experiments, the mice were inoculated intraperitoneally with 103 blood-derived trypomastigote forms, which had been obtained from previously infected mice.
Total RNA extraction and cDNA preparation by reverse-transcription (RT) PCR.
Total RNA was isolated from the cardiac tissue of mice by use of Trizol LS (Life Technologies) reagent, in accordance with the manufacturer's instructions. Briefly, the samples were homogenized, and 0.2 mL of chloroform (Sigma) was added to each 1 mL of Trizol LS reagent. The samples were then centrifuged at 12,000 g for 15 min at 4°C, and the aqueous phase was transferred to a clean tube. The same volume of isopropyl alcohol was added, and the samples were mixed in a vortex and incubated for 15 min at -20°C, to precipitate the RNA from the aqueous phase. After further centrifugation, the RNA pellets were washed in 75% ethanol, and the samples were suspended in water at a concentration of 0.5 g of RNA/L. cDNA was synthesized by use of Superscript II reverse transcriptase (Gibco).
Chemokine mRNA detection.
The expression of mRNA for chemokines (CCL3, CCL4, and CCL5) and -actin was analyzed by RT-PCR, which was performed with Taq polymerase (Gibco) in a PTC-100 thermal cycler (MJ Research). The reaction conditions were 35 cycles of 1 min at 94°C, 1 min at 54°C, and 2 min at 72°C, with a final extension step of 7 min at 72°C. For each set of primers, a negative sample (water) was run in parallel. The PCR products were separated by acrylamide gel electrophoresis and stained with silver nitrate. The primer sequences and PCR product sizes have been published elsewhere [9].
Spleen-cell cultures.
Suspensions of splenocytes from uninfected mice were washed in Hanks' balanced salt solution (HBSS) and treated with lysis buffer (9 parts 0.16 mol/L ammonium chloride and 1 part 0.17 mol/L Tris-HCl [pH 7.5]) for 4 min. The erythrocyte-free cells were then washed 3 times in HBSS and adjusted to 3 × 106 cells/mL of RPMI 1640 (Flow Laboratories) supplemented with 10% fetal calf serum (Hyclone), 2-mercaptoethanol (5 × 10-5 mol/L), L-glutamine (2 mmol/L), and antibiotics (all from Sigma). The cell suspension was distributed (1 mL/well) in 24-well tissue-culture plates (Corning) and cultured for 40 h at 37°C in a humidified 5% CO2 atmosphere, in the presence or absence of trypomastigote forms of T. cruzi (5 parasites/cell) or concanavalin A (ConA; 2 g/mL).
Isolation of inflammatory cells from cardiac tissue of T. cruziinfected mice.
To isolate mononuclear cells from myocardia, 1520 mouse hearts removed 17 days after infection were washed (to remove blood clots), pooled, minced with scissors into 12-mm fragments, extensively washed, and subjected to enzymatic digestion with a solution containing 0.05% trypsin (Gibco) and 0.01% collagenase type II (Worthington) for 10 min at 37°C. The cell suspension was spun, the supernatant was removed, and the pellet was suspended in 2 mL of 100% Percoll (Pharmacia). Aliquots (2 mL) of Percoll (70% and 40%) were added to the top of the cell suspension, and the suspension was centrifuged at 1500 g for 30 min. The leukocytes (between the 70% and 40% fractions) were removed and washed with RPMI 1640 (5%), and their phenotypes were evaluated by flow cytometry.
Flow-cytometry analysis.
Both inflammatory cells obtained from the cardiac tissue of T. cruziinfected mice and splenocytes were stained with fluorescein isothiocyanate (FITC) or phosphatidylethanolamine-labeled antibodies against CD3, CD4, CD8, CCR5 (Pharmingen), or CCR3 (Santa Cruz Biotechnology) molecules. Viable cells (3 × 105) were analyzed by use of a flow cytometer and CellQuest software (version 3.3; Becton Dickinson).
Histological evaluation.
Groups of 34 mice were killed on different days after infection with T. cruzi. Hearts were fixed in neutral 10% formalin, embedded in paraffin, sectioned, stained with hematoxylin-eosin, and examined by light microscopy.
Immunohistochemical analysis.
Cardiac tissues of mice were removed on days 17 and 20 after infection with T. cruzi, placed in compound-embedding medium (Sakura Finetek), snap-frozen by use of liquid nitrogen, and stored at -70°C. Ten-micrometer sections were prepared in a cryostat, collected on polylysine-coated slides, fixed with cold acetone, and allowed to air dry.
The endogenous peroxidase activity was blocked by use of 3% hydrogen peroxide for 20 min, and nonspecific binding was blocked by use of goat serum (diluted 1 : 200 in PBS) for 45 min. The slides were washed and incubated overnight with goat antimouse MIP-1, MIP-1, RANTES, or CCR5 (Santa Cruz Biotechnology) or with rat antimouse CD4 or CD8 (Pharmingen) diluted 100 times in PBS1% bovine serum albumin (BSA). After extensive washes and incubation for 30 min with biotin-labeled rabbit antigoat or goat antirat antibody (Pharmingen), the reaction product was detected by use of avidin-biotinperoxidase complex (Vector Laboratories), the color of the reaction was developed by use of diaminobenzidine tetrahydrochloride (DAB; Sigma), and the reaction was counterstained with Mayer's hematoxylin. Controls were performed by incubation of cells with nonimmune goat or rat IgG.
Immunofluorescent staining and confocal analysis.
Cryostat sections obtained and fixed as described above were incubated overnight at 4°C with antiCD16/CD32 monoclonal antibody (MAb; Pharmigen) in PBS1% BSA and then were blocked with avidin-biotin. Sections were incubated with the appropriate dilutions of rat antimouse MAb against CD4 and CD8 (labeled with FITC) and biotinylated anti-CCR5 or normal rat IgG for 1 h. Immunostaining for CCR5 was detected by incubation with streptavidinTexas red (Pharmigen) for 1 h. Between each incubation step, slides were washed 3 times in PBS and mounted in aqueous medium. The slides were analyzed by fluorescent microscopy (Leica), and the images were processed by use of Adobe Photoshop software (version 7.0).
Statistical analysis.
Results are expressed as the mean ± SE of the triplicate cultures or experiments. Statistical analysis was performed by use of analysis of variance, followed by the Student-Newman-Keuls test (InStat software; GraphPad). P < .05 was considered to be statistically significant.
RESULTS
Phenotype of inflammatory cells in the cardiac tissue of T. cruziinfected mice.
Because CCL3, CCL4, and CCL5 bind to CCR5 on the surface of leukocytes, we examined whether the inflammatory cells in the myocardia of infected mice expressed CCR5. These experiments showed that there were very few CD4+ and CD8+ T cells in uninfected mice (figure 2A) and that the number of these cells increased markedly after infection (figure 2C and 2D). When we examined mouse cardiac tissue obtained during the acute phase of infection, we found that the inflammatory infiltrate was predominantly composed of CD8+ T cells (figure 2C and 2D). The quantification of T cells infiltrating the cardiac tissue (figure 3A) revealed that the majority of CD3+ T cells in the cardiac tissue of infected mice were also CD8+ (figure 3B). Other cells found in cardiac tissue expressed CD11c (11%), CD14 (7%), and CD56 (20%). Expression of CCR5 was found to be positive in the majority of leukocytes in inflammatory infiltrate (figure 2B), indicating that a significant amount of CD8+ T cells expressed CCR5. Indeed, double immunofluorescence labeling revealed that the majority of CD8+ T cells (figure 2F), but not of CD4+ T cells (figure 2E), expressed CCR5. These data were confirmed by flow-cytometry analysis, which showed that 70% of CD8+ and 20% of CD4+ T cells that were isolated from myocardia during acute infection expressed CCR5 (figure 3A and 3B).
We found a linear increase in the expression of CCR5 on T cells after infection, on the basis that the increase was significant as soon as 5 days after infection (figure 4B). The increased expression of CCR5 was almost exclusively on CD8+ T cells; expression on CD4+ T cells was not significantly increased (figure 4B). These data indicate that T. cruzi is able to induce expression of CCR5 on CD8+ T splenocytes.
Greater susceptibility of CCR5-/- mice to infection with T. cruzi, which mediates the cell influx to cardiac tissue.
To investigate the relevance of CCR5 to T. cruzi infection, CCR5-/- mice were infected with T. cruzi. The results showed a significantly increased parasitemia during the acute phase of infection in CCR5-/- mice (between days 715 after infection) (figure 5A). Moreover, although the mortality of wt mice was 50% on day 26 after infection, all CCR5-/- had died by that day (P < .05) (figure 5B). Interestingly, the intensity and kinetics of diffuse and focal inflammatory infiltrates were different in the cardiac tissue of CCR5-/- and wt mice infected with the parasite. Although, on day 20 after infection, significant inflammatory infiltrates were found in the myocardia of wt mice infected with T. cruzi (figure 6C), these were almost absent in the myocardia of CCR5-/- mice infected with T. cruzi (figure 6D). Instead, we found very large amounts of parasite nests (figure 6D), such that in several microscopy fields the amounts of parasites in the tissue were virtually uncountable. Also noticeable was the finding that the sizes of parasite nests in CCR5-/- mice were bigger than those in wt mice. In contrast, rare, isolated parasite nests were found in the cardiac tissue of wt mice (figure 6C). Absence of cell infiltrate was found in uninfected wt (figure 6A) and CCR5-/- (figure 6B) mice.
DISCUSSION
It is now clear that IFN- and TNF-activated macrophages control of the replication of T. cruzi in various hosts [20]. The mechanism by which macrophages are able to kill intracellular parasites has been shown to be dependent on NO [813, 20], although a recent study using T. cruzi strains Tulahuen and Brazil suggested that inducible NO synthase (iNOS) is not required to control parasite replication in vivo [21]. However, much less is known about the mechanisms governing the migration of leukocytes to sites of tissue infection, especially to hearts of infected hosts. More recently, a group of mediators, the chemokines, have been described in some detail and have been shown to participate in the recruitment of leukocytes in several models of inflammatory diseases [14, 2224]. In the present study, we demonstrate an important role for CCR5-acting chemokines in the control of leukocyte migration and parasite replication during the acute phase of infection with the Y strain of T. cruzi. This strain induces a parasitemia that peaks around days 911 of infection and causes an intense inflammatory reaction in cardiac tissue that is more intense between days 13 and 20 [25].
Infection of macrophages [9] and cardiomyocytes [10] with T. cruzi trypomastigote forms is accompanied by the significant production of several chemokines, including CCL3, CCL4, and CCL5. Not only can the latter cells produce chemokines, but chemokines can also act on the cells to induce iNOS expression, NO expression, and the killing of parasites [913]. Thus, in the present study, it was important to ascertain whether chemokine expression would also occur in vivo after infection of mice with the Y strain of T. cruzi. Indeed, both mRNA and protein for CCL3, CCL4, and CCL5 could be found in the cardiac tissue of T. cruziinfected mice, in close correlation with tissue inflammation; these results are in agreement with the findings of other studies [1416]. Notably, these latter studies showed that the expression of chemokines was greatly under the control of IFN- and that the modulation of chemokine expression was accompanied by the modulation of the recruitment of leukocytes to sites of T. cruziinduced inflammation [26]. Altogether, these studies and the present data demonstrate the close correlation between the expression of chemokines and the development of tissue inflammation after T. cruzi infection in vivo.
Not only was the expression of chemokines enhanced, but the expression of CCR5, a receptor for CCL3, CCL4, and CCL5, was also enhanced on infiltrating leukocytes in the hearts of T. cruziinfected mice. More specifically, there was a marked increase in the number of CD8+ T cells expressing CCR5, as assessed by both immunohistochemistry and flow-cytometry analysis. The number of CD4+ T cells expressing CCR5 in the cardiac tissue of infected mice also increased, but to a lesser extent. Interestingly, the increased expression of CCR5 on tissue lymphocytes was mirrored by a significant increase in the expression of this receptor on circulating lymphocytes. Indeed, infection with T. cruzi, but not stimulation with ConA, also enhanced the expression of CCR5 on both CD4+ and CD8+ T cells cultivated in vitro. These results are in agreement with those of a recent study that showed that both CD3+CD4+ and CD3+CD8+ T cells obtained from the peripheral blood of infected patients expressed CCR5 to a greater degree than did those obtained from uninfected individuals [27]. In the latter study, the enhanced CCR5 expression on T cells was lost only when heart failure supervened chronic Chagas disease. Thus, infection with T. cruzi was accompanied by a parasite-associated increase in the expression of CCR5 and migration of T cells to infected tissues, in which several CCR5-acting chemokines are produced. The increased expression of CCR5 could be due to the production of IFN- [28, 29], TNF- [8, 30], interleukin (IL)10 [7, 31], and/or IL-12 [30, 32, 33] that occurs after the addition of trypomastigote forms of T. cruzi to cultures of splenocytes or after the infection of mice. Indeed, CCR5 expression is positively regulated by these cytokines [3437].
The role of CCR5 in the control of T. cruzi infection and in the disease associated with the infection was evaluated in CCR5-/- mice. In these mice, a higher level of parasitemia and greater mortality was observed, compared with those in T. cruziinfected wt mice. Moreover, the amount of parasites in the cardiac tissue of CCR5-/- mice was greatly enhanced. The reduced control of parasite replication was associated with a dramatic reduction of the migration of T cells and overall disease, indicating that CCR5 was crucial to the migration of lymphocytes to the hearts of infected mice. Importantly, there was no defect in the ability of macrophages from CCR5-/- mice to produce NO and IL-12 or to kill the parasite in vitro (data not shown). Similar to our results in intracellular adhesion molecule1deficient mice [38], the decreased number of lymphocytes in cardiac tissue was associated with decreased local production of IFN- (data not shown) and an increased level of parasitemia, suggesting that there was a defect in the migration of cells capable of producing cytokines (such as IFN-) that activate the production of NO. Thus, the lack of CCR5 is accompanied by the inability of CCR5+ T cells to migrate to the hearts of infected mice and the inability of the host to control parasite replication. Similarly, absence of CCR5 leads to increased susceptibility to Cryptococcus neoformans infection, because of the absence of the migration of cells to the brain [18]. On the contrary, a defective trafficking of leukocytes in CCR5-/- mice has been associated with (1) resistance to the development of cerebral malaria in mice infected with Plasmodium berghei [39] and (2) demyelination after infection with mouse hepatitis virus [40].
CD8+ T cells compose the great majority of cells observed in the inflammatory infiltrate in the hearts of patients with chagasic cardiomyopathy [4144]. These cells may play a role in the damage to the heart that is observed during the chronic phase of disease [4144]. Indeed, cardiac lesions observed during the chronic phase of infection with T. cruzi have been associated with the local inflammatory response that is driven by host immune cells [45, 46]. It is not known, however, whether the CD8+ T cells that infiltrate the hearts of patients with Chagas disease also express CCR5 or whether there is a functional relevance for the observed increase of CCR5 expression on the CD8+ T cells that are obtained from the peripheral blood of patients with Chagas disease [27]. A role for CCR5 in human disease has also been suggested by a study that showed that a point mutation of the CCR5 promoter was significantly increased in asymptomatic patients, compared with that in patients with chagasic cardiomyopathy [47]. It is clear that further studies are necessary to understand the role played by CCR5 in the pathogenesis of Chagas disease.
In summary, our results demonstrate that CCR5-acting chemokines are expressed in the hearts of T. cruziinfected mice and that CCR5 is most relevant to the control of the lymphocyte migration and parasite replication. Future studies should be conducted to evaluate whether CCR5 is also relevant to the pathophysiology of chronic infection and whether the blockade of CCR5 could be a viable target for the prevention of the migration of lymphocytes to inflamed cardiac tissue.
References
1. Russo M, Starobinas N, Minoprio P, Coutinho A, Hontebeyrie-Joskowicz M. Parasitic load increases and myocardial inflammation decreases in Trypanosoma cruziinfected mice after inactivation of helper T cells. Ann Inst Pasteur Immunol 1988; 139:22536. First citation in article
2. Rottenberg ME, Riarte A, Sporrong L, et al. Outcome of infection with different strains of Trypanosoma cruzi in mice lacking CD4 and/or CD8. Immunol Lett 1995; 45:5360. First citation in article
3. Tarleton RL, Grusby MJ, Postan M, Glimcher LH. Trypanosoma cruzi infection in MHC-deficient mice: further evidence for the role of both class I and class IIrestricted T cells in immune resistance and disease. Int Immunol 1996; 8:1322. First citation in article
4. Tarleton RL, Koller BH, Latour A, Postan M. Susceptibility of 2-microglobulindeficient mice to Trypanosoma cruzi infection. Nature 1992; 356:33840. First citation in article
5. McCabe RE, Meagher SG, Mullins BT. Endogenous interferon-, macrophage activation, and murine host defense against acute infection with Trypanosoma cruzi. J Infect Dis 1991; 163:9125. First citation in article
6. Torrico F, Heremans H, Rivera MT, Van Marck E, Billiau A, Carlier Y. Endogenous IFN- is required for resistance to acute Trypanosoma cruzi infection in mice. J Immunol 1991; 146:362632. First citation in article
7. Silva JS, Morrissey PJ, Grabstein KH, Mohler KM, Anderson D, Reed SG. Interleukin 10 and interferon regulation of experimental Trypanosoma cruzi infection. J Exp Med 1992; 175:16974. First citation in article
8. Silva JS, Vespa GN, Cardoso MA, Aliberti JC, Cunha FQ. Tumor necrosis factor mediates resistance to Trypanosoma cruzi infection in mice by inducing nitric oxide production in infected interferonactivated macrophages. Infect Immun 1995; 63:48627. First citation in article
9. Aliberti JC, Machado FS, Souto JT, et al. -Chemokines enhance parasite uptake and promote nitric oxidedependent microbiostatic activity in murine inflammatory macrophages infected with Trypanosoma cruzi. Infect Immun 1999; 67:481926. First citation in article
10. Machado FS, Martins GA, Aliberti JC, Mestriner FL, Cunha FQ, Silva JS. Trypanosoma cruziinfected cardiomyocytes produce chemokines and cytokines that trigger potent nitric oxidedependent trypanocidal activity. Circulation 2000; 102:30038. First citation in article
11. Vespa GN, Cunha FQ, Silva JS. Nitric oxide is involved in control of Trypanosoma cruziinduced parasitemia and directly kills the parasite in vitro. Infect Immun 1994; 62:517782. First citation in article
12. Lima MF, Zhang Y, Villalta F. -Chemokines that inhibit HIV-1 infection of human macrophages stimulate uptake and promote destruction of Trypanosoma cruzi by human macrophages. Cell Mol Biol (Noisy-le-grand) 1997; 43:106776. First citation in article
13. Villalta F, Zhang Y, Bibb KE, Kappes JC, Lima MF. The cysteine-cysteine family of chemokines RANTES, MIP-1, and MIP-1 induce trypanocidal activity in human macrophages via nitric oxide. Infect Immun 1998; 66:46905. First citation in article
14. Aliberti JC, Souto JT, Marino AP, at el. Modulation of chemokine production and inflammatory responses in interferon- and tumor necrosis factorR1deficient mice during Trypanosoma cruzi infection. Am J Pathol 2001; 158:143340. First citation in article
15. Talvani A, Ribeiro CS, Aliberti JC, et al. Kinetics of cytokine gene expression in experimental chagasic cardiomyopathy: tissue parasitism and endogenous IFN- as important determinants of chemokine mRNA expression during infection with Trypanosoma cruzi. Microbes Infect 2000; 2:85166. First citation in article
16. dos Santos PV, Roffe E, Santiago HC, et al. Prevalence of CD8+ T cells in Trypanosoma cruzielicited myocarditis is associated with acquisition of CD62LLowLFA-1HighVLA-4High activation phenotype and expression of IFN-inducible adhesion and chemoattractant molecules. Microbes Infect 2001; 3:97184. First citation in article
17. Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol 2004; 22:891928. First citation in article
18. Huffnagle GB, McNeil LK, McDonald RA, et al. Cutting edge: role of C-C chemokine receptor 5 in organ-specific and innate immunity to Cryptococcus neoformans. J Immunol 1999; 163:46426. First citation in article
19. Zhou Y, Kurihara T, Ryseck RP, et al. Impaired macrophage function and enhanced T celldependent immune response in mice lacking CCR5, the mouse homologue of the major HIV-1 coreceptor. J Immunol 1998; 160:401825. First citation in article
20. Silva JS, Machado FS, Martins GA. The role of nitric oxide in the pathogenesis of Chagas disease. Front Biosci 2003; 8:31425. First citation in article
21. Cummings KL, Tarleton RL. Inducible nitric oxide synthase is not essential for control of Trypanosoma cruzi infection in mice. Infect Immun 2004; 72:40819. First citation in article
22. Souto JT, Aliberti JC, Campanelli AP, et al. Chemokine production and leukocyte recruitment to the lungs of Paracoccidioides brasiliensisinfected mice is modulated by interferon-. Am J Pathol 2003; 163:58390. First citation in article
23. Ji J, Sun J, Soong L. Impaired expression of inflammatory cytokines and chemokines at early stages of infection with Leishmania amazonensis. Infect Immun 2003; 71:427888. First citation in article
24. Godessart N, Kunkel SL. Chemokines in autoimmune disease. Curr Opin Immunol 2001; 13:6705. First citation in article
25. Silva JS, Rossi MA. Intensification of acute Trypanosoma cruzi myocarditis in BALB/c mice pretreated with low doses of cyclophosphamide or irradiation. J Exp Pathol (Oxford) 1990; 71:339. First citation in article
26. Teixeira MM, Gazzinelli RT, Silva JS. Chemokines, inflammation and Trypanosoma cruzi infection. Trends Parasitol 2002; 18:2625. First citation in article
27. Talvani A, Rocha MO, Ribeiro AL, Correa-Oliveira R, Teixeira MM. Chemokine receptor expression on the surface of peripheral blood mononuclear cells in Chagas disease. J Infect Dis 2004; 189:21420. First citation in article
28. Cardillo F, Voltarelli JC, Reed SG, Silva JS. Regulation of Trypanosoma cruzi infection in mice by interferon and interleukin 10: role of NK cells. Infect Immun 1996; 64:12834. First citation in article
29. Graefe SE, Jacobs T, Gaworski I, Klauenberg U, Steeg C, Fleischer B. Interleukin-12 but not interleukin-18 is required for immunity to Trypanosoma cruzi in mice. Microbes Infect 2003; 5:8339. First citation in article
30. Hunter CA, Slifer T, Araujo F. Interleukin-12mediated resistance to Trypanosoma cruzi is dependent on tumor necrosis factor alpha and gamma interferon. Infect Immun 1996; 64:23816. First citation in article
31. Holscher C, Mohrs M, Dai WJ, et al. Tumor necrosis factor mediated toxic shock in Trypanosoma cruziinfected interleukin 10deficient mice. Infect Immun 2000; 68:407583. First citation in article
32. Aliberti JC, Cardoso MA, Martins GA, Gazzinelli RT, Vieira LQ, Silva JS. Interleukin-12 mediates resistance to Trypanosoma cruzi in mice and is produced by murine macrophages in response to live trypomastigotes. Infect Immun 1996; 64:19617. First citation in article
33. Antunez MI, Cardoni RL. IL-12 and IFN- production, and NK cell activity, in acute and chronic experimental Trypanosoma cruzi infections. Immunol Lett 2000; 71:1039. First citation in article
34. Croitoru-Lamoury J, Guillemin GJ, Boussin FD, et al. Expression of chemokines and their receptors in human and simian astrocytes: evidence for a central role of TNF and IFN in CXCR4 and CCR5 modulation. Glia 2003; 41:35470. First citation in article
35. Takayama T, Morelli AE, Onai N, et al. Mammalian and viral IL-10 enhance C-C chemokine receptor 5 but down-regulate C-C chemokine receptor 7 expression by myeloid dendritic cells: impact on chemotactic responses and in vivo homing ability. J Immunol 2001; 166:713643. First citation in article
36. Iwasaki M, Mukai T, Nakajima C, et al. A mandatory role for STAT4 in IL-12 induction of mouse T cell CCR5. J Immunol 2001; 167:687783. First citation in article
37. Juffermans NP, Paxton WA, Dekkers PE, et al. Up-regulation of HIV coreceptors CXCR4 and CCR5 on CD4+ T cells during human endotoxemia and after stimulation with (myco)bacterial antigens: the role of cytokines. Blood 2000; 96:264954. First citation in article
38. Michailowsky V, Celes MRN, Marino AP, et al. ICAM-1 deficiency leads to impaired recruitment of T lymphocytes and enhanced host susceptibility to infection with Trypanosoma cruzi. J Immunol 2004; 173:46370. First citation in article
39. Belnoue E, Kayibanda M, Deschemin JC, et al. CCR5 deficiency decreases susceptibility to experimental cerebral malaria. Blood 2003; 101:42539. First citation in article
40. Glass WG, Liu MT, Kuziel WA, Lane TE. Reduced macrophage infiltration and demyelination in mice lacking the chemokine receptor CCR5 following infection with a neurotropic coronavirus. Virology 2001; 288:817. First citation in article
41. Higuchi Mde L, Gutierrez PS, Aiello VD, et al. Immunohistochemical characterization of infiltrating cells in human chronic chagasic myocarditis: comparison with myocardial rejection process. Virchows Arch A Pathol Anat Histopathol 1993; 423:15760. First citation in article
42. Reis MM, Higuchi Mde L, Benvenuti LA, et al. An in situ quantitative immunohistochemical study of cytokines and IL-2R+ in chronic human chagasic myocarditis: correlation with the presence of myocardial Trypanosoma cruzi antigens. Clin Immunol Immunopathol 1997; 83:16572. First citation in article
43. Reis DD, Jones EM, Tostes S Jr, et al. Characterization of inflammatory infiltrates in chronic chagasic myocardial lesions: presence of tumor necrosis factor+ cells and dominance of granzyme A+, CD8+ lymphocytes. Am J Trop Med Hyg 1993; 48:63744. First citation in article
44. Higuchi MD, Ries MM, Aiello VD, et al. Association of an increase in CD8+ T cells with the presence of Trypanosoma cruzi antigens in chronic, human, chagasic myocarditis. Am J Trop Med Hyg 1997; 56:4859. First citation in article
45. Brener Z, Gazzinelli RT. Immunological control of Trypanosoma cruzi infection and pathogenesis of Chagas' disease. Int Arch Allergy Immunol 1997; 114:10310. First citation in article
46. Abel LC, Rizzo LV, Ianni B, et al. Chronic Chagas' disease cardiomyopathy patients display an increased IFN- response to Trypanosoma cruzi infection. J Autoimmun 2001; 17:99107. First citation in article
47. Calzada JE, Nieto A, Beraun Y, Martin J. Chemokine receptor CCR5 polymorphisms and Chagas' disease cardiomyopathy. Tissue Antigens 2001; 58:1548. First citation in article