Down-regulation of host immune response to Toxoplasma gondii is associated with the expression of specific cytokines, in particular IL-10, and the induction of CD4+ T cell anergy. In the present study we report that the expression of both CD4 and CD2 antigen is down-regulated during the acute phase of infection. A decrease in the expression of CD2 was apparent during the acute phase of T. gondii infection in three genetically distinct strains of mice, CBA/J, C57BL/6, and BALB/c. The lymphoproliferative response induced by cross-linked anti-CD3 mAb or by Con A was markedly depressed. This suppressed response was associated with a reduction in the influx of Ca2+. We have examined whether lymphocytes from T. gondii mice maintain NF-AT transcription factors in the nucleus where they participate in the Ca2+-dependent induction of genes required for lymphocyte activation and proliferation. Immunofluorescence with confocal microscopy using an Ab to NF-ATc demonstrates a decrease in translocation of NF-ATc in T lymphocytes from acutely infected mice. Together, these results suggest that the defect in T cell expansion that occurs during acute murine toxoplasmosis is related to reduced activity of NF-AT, a calcium-dependent transcription factor required for T cell proliferation.

The intracellular protozoan parasite Toxoplasma gondii is a major opportunistic pathogen of the newborn and those with AIDS. It has been demonstrated that suppression of lymphocyte proliferation occurs in response to parasite Ag and mitogen during acute toxoplasmosis in both humans and mice (1, 2, 3). In humans this down-regulatory response can be mediated in vitro by parasite-infected monocytes (4). In mice, both spleen-derived macrophages and T cells can elaborate this suppressive response (5, 6, 7).

A number of soluble cell products has been implicated in this down-regulation. In mice, the maximal immune suppression (day 7 postinfection) has been associated with a significant reduction in IL-2 secretion (6). In vitro, the immunosuppressive effect on IL-2 production is dependent upon the number of parasites used to infect macrophages (8). Both IL-10 and nitric oxide appear to play a role in manipulating the down-regulatory event. In addition to the well-recognized role for IFN-γ in host resistance to this parasite (9, 10, 11), in humans and perhaps mice this cytokine appears to partially mediate the release of an immune down-regulatory soluble factor (4). Acute toxoplasma infection in mice can induce a state of T cell unresponsiveness (12, 13). On day 7 postinfection, a partial reduction in the proliferative response of all CD4+ T cells to mitogen or parasite Ag stimulation was observed, particularly in Vb5 cells. Addition of rIL-2 partially restored the CD4+ T cell proliferative response in vitro. These studies suggested that the activation-induced CD4+ T cell unresponsiveness may be an important immune down-regulatory event in the infected host (12, 13).

It has been observed that after T cell activation rapid changes in the function or expression of several membrane-associated molecules occur, including CD11a, CD2, and CD69 (14, 15). Activated T cells require a primary signal mediated via triggering of the Ag-specific TCR and a secondary accessory signal, such as CD2, CD4, CD8, and CD28 (16). It has been shown that anti-CD2 mAbs can block T cell activation in vitro, implying an important role for CD2 in T cell activation (17). In T lymphocytes, the binding of mAb to the CD3 complex mimics activation via the Ag receptor, resulting in the production of inositol 1,4,5-trisphosphate, an increased intracellular ionized Ca+ concentration, and subsequent proliferation (18). Some microbial pathogens, including HIV, Trypanosome cruzi, and Leishmania donovani (19, 20, 21) have been associated with the defective regulation of [Ca+]i3 in response to extracellular stimuli. The calcium-mediated signaling event is essential for growth, death, differentiation, and function of immune cells (22, 23). Several Ca2+-sensitive transcriptional regulators, NF-κB, JNK, and NF-AT, participate in the expression of genes that underlie these responses (24). Sustained high concentrations of Ca+ are required to maintain NF-AT transcription factors in the nucleus, where they participate in Ca2+-dependent induction of the genes required for lymphocyte activation and proliferation (25).

In this study we report that acute infection with toxoplasma in mice is able to alter the expression of several T cell membrane molecules, in particular CD2 and CD4. Furthermore, we observed that acute infection with this obligate intracellular parasite alters both the CD3-activated and mitogen-induced [Ca+]i response. The effect of this response on the Ca2+-sensitive transcriptional regulator NF-AT was determined during acute murine infection with this parasite.

Female CBA/J (H-2k), BALB/c (H-2d), and C57BL/6 (H-2b) mice, 5–6 wk old, were purchased from The Jackson Laboratory (Bar Harbor, ME). All animals were housed in the accredited Animal Research Facility at Dartmouth Medical School (Hanover, NH) and maintained under the guidelines established by the institution for their use. The parental P strain of T. gondii (PLK) was used for our experiments. Parasites were maintained in our laboratory by in vitro passage in human foreskin fibroblasts at 37°C in MEM medium without calf serum. Parasites were purified from human fibroblast cell culture as previously described (6). Each mouse received 1.2 × 105 tachyzoites/i.p. injection.

Mice were killed on day 5 after infection, and spleens were removed and gently dissociated into single cell suspensions. RBCs were removed using lysing buffer (Sigma, St. Louis, MO). Cell suspensions were passed through nylon wool columns to enrich for T cells. These cells were >90% T cells. The cells were stained, and flow cytometric analysis was performed. T cell proliferation assays was performed as previously described (6, 26). Briefly, lymphocytes (2 × 105) were suspended in 200 μl of complete medium and cultured in 96-well microtiter plates in the presence or the absence of Con A and cross-linked anti-CD3 mAb. For the anti-CD3 mAb (PharMingen, San Diego, CA; 145-2C11 mAb)-driven proliferation assay, culture plate wells were precoated with goat anti-hamster IgG (14 μg of anti-hamster IgG; Jackson Immunology Research Laboratories, West Grove, PA) overnight at 4°C. After washing, the wells were incubated with different concentrations of anti-CD3 mAb at 37°C for several hours. Splenocytes were added to the wells and were cultured, and DNA synthesis was determined after 42 h by 6-h thymidine incorporation of [methyl-3H]thymidine (ICN, Costa Mesa, CA). All cell culture and FACS analysis experiments were performed in 96-well plates.

Abs directed against CD2 (mAb RM2-5, PE-conjugated), CD3 (mAb 145-2C11, FITC-conjugated), CD4 (mAb RM4.5, FITC-conjugated), CD8 (mAb 53-6.7, FITC-conjugated), and TCR (mAb H57-597, FITC-conjugated) were used in this experiment. All Abs directed against these epitopes were purchased from PharMingen (San Diego, CA). Direct immunofluorescence PE- or FITC-conjugated mAb was added to 1 × 106 cells, followed by a 45-min incubation on ice. Cells were then washed twice with PBS containing 1% BSA and fixed in 1% formaldehyde. Negative controls were stained with PE- or FITC-conjugated mouse Ig.

Spleen cells were obtained from mice on day 5 postinfection and from uninfected mice. Cell suspensions were passed through nylon wool columns to enrich for T cells. Changes in Ca+ were measured by flow cytometry using indo-1/AM (Molecular Probes, Eugene, OR) on a HH/2150 flow cytometer (FACStar, Becton Dickinson, Mountain View, CA) as previously described (27). Briefly, after lysis of RBCs, cells were washed with serum-free medium and loaded with 5 μM indo-1 (Molecular Probes) for 45 min at 37°C. For each assay, indo-1-loaded cells were diluted to 1 × 105/ml with medium containing 5% FCS, equilibrated at 37°C, and analyzed by flow cytometry. All unstimulated cells were removed before analysis. Con A or anti-CD3 mAb was added 30 s before the beginning of the experiment. To analyze CD4-positive cells, lymphocytes were loaded with indo-1/AM and stained with FITC-conjugated CD8 mAb (18), and then FITC (CD8+) fluorescent CD8+ cells were excluded from analysis by electronic gating (18). The cells were analyzed at about 250 cells/s by means of dual-laser FACS. The calcium concentration was determined by the ratio between 485 nm and 405 nm emission with 355 nm excitation. Calibration was performed by measuring Rmin and Rmax in cells, and applying the equation described previously (28). Responses are reported as Ca+ concentrations vs time.

For this assay, the Ab 7A6 (anti NF-ATc) were provided by Dr. Luika Timmerman (Stanford University School of Medicine, Palo Alto, CA). Cells from uninfected and day 5 postinfection animals were stimulated with ionomycin and PMA (1 μM and 10 ng/ml, respectively; for 20 min). All cells were prepared as described previously (25). Briefly, after centrifugation (Cytospin, Shandon, Pittsburgh, PA; 3 min at 300 rpm), cells were fixed in 4% paraformaldehyde in PBS for 10 min, permeabilized with 0.1% Triton X-100 in PBS for 10 min at room temperature, and rehydrated in PBS. The cells were then incubated overnight at 4°C with NF-AT-specific mAb 7A6 (1/500 in PBS) followed by anti-mouse biotin-conjugated (1/1000; Sigma) and avidin-FITC-conjugated (1/200; PharMingen, CA) Abs.

Levels of significance of the differences between groups were determined by Student’s t test. Statistical significance was set at p < 0.05 for all comparisons.

The cells from infected mice were assayed for their ability to initiate DNA synthesis in response to either CD3-mediated activation or mitogen-driven stimulation. For these experiments, spleen cells from mice infected with T. gondii for 5 days were cultured with either anti-CD3 mAb or Con A. Splenocytes from infected mice failed to proliferate in response to either anti-CD3 mAb or mitogen stimulation compared with those from control mice (Table I; p < 0.005). As previously reported, the addition of rIL-2 to the suppressed splenoyctes from infected mice increased the proliferative response to several stimuli, including mitogen, parasite Ag, and live parasites (data not shown) (6, 8).

Table I.

Lymphocytes responses to different concentrations of cross-linked anti-CD3 mAb or Con Aa

Stimulus[3H]Thymidine Incorporation (cpm)
UninfectedbT. gondii infectedc
CD3 mAb/ml   
5.0 μg 122,520 ± 1,969 24,592 ± 654 
2.0 μg 96,250 ± 5,449 11,877 ± 1,583 
0.5 μg 77,365 ± 2,781 4,930 ± 245 
ConA/ml   
5.0 μg 106,850 ± 3,039 13,575 ± 77 
2.0 μg 74,299 ± 133 9,521 ± 77 
0.5 μg 35,575 ± 1,376 5,286 ± 457 
Stimulus[3H]Thymidine Incorporation (cpm)
UninfectedbT. gondii infectedc
CD3 mAb/ml   
5.0 μg 122,520 ± 1,969 24,592 ± 654 
2.0 μg 96,250 ± 5,449 11,877 ± 1,583 
0.5 μg 77,365 ± 2,781 4,930 ± 245 
ConA/ml   
5.0 μg 106,850 ± 3,039 13,575 ± 77 
2.0 μg 74,299 ± 133 9,521 ± 77 
0.5 μg 35,575 ± 1,376 5,286 ± 457 
a

Proliferative response to cross-linked anti-CD3 mAb or Con A of lymphocytes from CBA/J mice (n = 3/group) infected with T. gondii (PLK) tachyzoites (1.2 × 105/mouse) or age-matched uninfected mice. On day 5 after infection, lymphocytes from spleens of infected mice or control mice were cultured in quadruplicate (2 × 105 viable cells per well) in flat-bottom wells in RPMI 1640 plus 10% FCS. The cells were cultured in the presence of different concentrations of either anti-CD3 mAb (5, 2, and 0.5 μg/ml) or Con A (5, 2, and 0.5 μg/ml). Cells from uninfected and infected animals were cultured in medium only, and cpm in uninfected group is 2142 ± 145 and experimental group is 2250 ± 280. [3H]Thymidine incorporation was determined on day 3 of culture. The results are expressed as means ± SD for quadruplicate. These results are representative of three separate experiments.

b,c Significant difference in the levels of lymphoproliferation between uninfected (b) and infected mice (c) (b vs c, p < 0.005).

To determine the effect of T. gondii infection on the expression of T cell surface molecules, lymphocytes were isolated from day 5 postinfection CBA/J, BALB/c, and C57BL/6 mice, and the expression of T cell phenotypes was analyzed by FACS. As shown in Table II, a decrease in the expression of both CD2 and CD4 surface molecules was observed in all three strains of mice on day 5 postinfection. Compared with uninfected mice, this diminution in CD2 (p < 0.05) and CD4 (p < 0.005) molecules was statistically significant in all three strains of mice infected with T. gondii. The diminution in CD2 expression ranged from 11% (CBA/J) to 24% (C57BL/6). For CD4, the decrease in the number of cells expressing this phenotype fell approximately 27 ± 2% for all three strains of mice. There was no significant change in the expression of CD8+, although a modest, but insignificant, rise in the CBA/J and BALB/c mice was found. There was no difference in the expression of the αβ TCR molecules in cells from any of the three strains of mice infected with T. gondii.

Table II.

Phenotypic analysis of mouse T cell populationsa

Cell-Surface Ag ExpressionPercent Positive (mean fluorescence intensity)
Cells from uninfected miceCells from day 5 postinfected mice
CBA/JBALB/cC57BL/6CBA/JBALB/cC57BL/6
CD2 92 ± 4.0 (345 ± 7.0) 99 ± 1.5 (270 ± 2.6) 92 ± 2.0 (363 ± 3.0) 82 ± 3.5 (290 ± 8.0)b 87 ± 2.0 (145 ± 30)b 70 ± 3.0 (265 ± 1.5)b 
CD3 44 ± 1.0 (147 ± 1.4) 44 ± 1.0 (254 ± 2.0) 42 ± 1.0 (94 ± 4.5) 42 ± 3.0 (189 ± 10) 41 ± 2.0 (213 ± 10.0) 39 ± 1.0 (108 ± 3.0) 
CD4 23 ± 1.0 (515 ± 10.0) 29 ± 1.0 (344 ± 3.0) 24 ± 2.0 (138 ± 4.0) 17 ± 1.5 (386 ± 3.0)b 21 ± 1.0 (251 ± 2.5)b 17 ± 1.0 (98 ± 2.0)b 
CD8 13 ± 0.5 (235 ± 2.5) 10 ± 1.0 (476 ± 1.0) 14 ± 1.0 (192 ± 1.5) 15 ± 0.6 (162 ± 5) 12 ± 1.0 (270 ± 50) 8 ± 2.0 (135 ± 1.5) 
TCRαβ 29 ± 6.0 (80 ± 4.0) 40 ± 4.0 (203 ± 5.0) ND 35 ± 2.0 (62 ± 5.0) 32 ± 3.0 (165 ± 10.0) ND 
Cell-Surface Ag ExpressionPercent Positive (mean fluorescence intensity)
Cells from uninfected miceCells from day 5 postinfected mice
CBA/JBALB/cC57BL/6CBA/JBALB/cC57BL/6
CD2 92 ± 4.0 (345 ± 7.0) 99 ± 1.5 (270 ± 2.6) 92 ± 2.0 (363 ± 3.0) 82 ± 3.5 (290 ± 8.0)b 87 ± 2.0 (145 ± 30)b 70 ± 3.0 (265 ± 1.5)b 
CD3 44 ± 1.0 (147 ± 1.4) 44 ± 1.0 (254 ± 2.0) 42 ± 1.0 (94 ± 4.5) 42 ± 3.0 (189 ± 10) 41 ± 2.0 (213 ± 10.0) 39 ± 1.0 (108 ± 3.0) 
CD4 23 ± 1.0 (515 ± 10.0) 29 ± 1.0 (344 ± 3.0) 24 ± 2.0 (138 ± 4.0) 17 ± 1.5 (386 ± 3.0)b 21 ± 1.0 (251 ± 2.5)b 17 ± 1.0 (98 ± 2.0)b 
CD8 13 ± 0.5 (235 ± 2.5) 10 ± 1.0 (476 ± 1.0) 14 ± 1.0 (192 ± 1.5) 15 ± 0.6 (162 ± 5) 12 ± 1.0 (270 ± 50) 8 ± 2.0 (135 ± 1.5) 
TCRαβ 29 ± 6.0 (80 ± 4.0) 40 ± 4.0 (203 ± 5.0) ND 35 ± 2.0 (62 ± 5.0) 32 ± 3.0 (165 ± 10.0) ND 
a

Splenocytes were freshly obtained from mice (n = 2/group) infected for 5 days and cell-surface Ag expression was analyzed by FACS. The mean fluorescence intensity of the cell-surface Ag and the percentage of positive cells of 5000 cells analyzed were monitored with FITC or PE-conjugated mAbs and flow cytometry. Similar results were obtained in three repeated independent experiments.

b

, Indicates satistically significant difference in percentage or in mean fluorescence intensity (p < 0.05) between cells derived from infected mice vs uninfected mice.

Splenocytes from infected mice were analyzed for calcium mobilization using the fluorescence indicator indo-1 in conjunction with flow cytometry following CD3-mediated activation or mitogen stimulation. Cells that were stimulated with either anti-CD3 mAb or Con A had an impaired [Ca2+]i response compared with the control cells. In the response to anti-CD3 mAb (10 μg/ml) exposure, the [Ca2+]i achieved a maximum level of 850 nM (Fig. 1,A, uninfected) in cells from uninfected CBA/J mice. In contrast, the [Ca2+]i of lymphocytes from infected CBA/J mice on day 5 postinfection reached 450 nM (Fig. 1,A, LP infected). Similar results were obtained when the splenocytes from either infected or uninfected mice were stimulated with the T cell mitogen, Con A (Fig. 1,B). There was only a nominal reduction compared with control cells when [Ca2+]i mobilization was determined for splenocytes on day 2 postinfection (not shown). The difference in the degree of [Ca2+]i mobilization between anti-CD3 mAb activation and mitogen stimulation for both infected and uninfected conditions appears insignificant. Of note, 35% of the control cells responded to anti-CD3 mAb stimulation, whereas only 12% of the cells from T. gondii-infected mice demonstrated a shift in [Ca2+]i flux (data not shown). Parasite Ag was also used to assess the effect on [Ca2+]i mobilization. In this assay, mice were treated with formalin-fixed parasites (FFP; 5 × 105 parasites/mouse). Five days later their splenocytes were isolated and stimulated with either anti-CD3 mAb or Con A. As shown in Fig. 1 (A and B, FFP infected), there was no significant reduction of the [Ca2+]i mobilization in response to either stimulant following immunization with parasite Ag.

FIGURE 1.

Effects of CD3 mAb (A) or Con A (B) stimulation on [Ca2+]i of lymphocytes obtained from uninfected, FFP, or LP mice. Lymphocytes from uninfected, FFP infected (3 × 105), or LP infected (1.2 × 105) mice were loaded with indo-1 (45 min) and stimulated with anti-CD3 mAb (10 μg/ml) or Con A (10 μg/ml). CD3 mAb or Con A was added 30 s before the beginning of the experiment, and the cells were analyzed at 250 cells/s. Results are plotted as the mean calcium concentration vs time.

FIGURE 1.

Effects of CD3 mAb (A) or Con A (B) stimulation on [Ca2+]i of lymphocytes obtained from uninfected, FFP, or LP mice. Lymphocytes from uninfected, FFP infected (3 × 105), or LP infected (1.2 × 105) mice were loaded with indo-1 (45 min) and stimulated with anti-CD3 mAb (10 μg/ml) or Con A (10 μg/ml). CD3 mAb or Con A was added 30 s before the beginning of the experiment, and the cells were analyzed at 250 cells/s. Results are plotted as the mean calcium concentration vs time.

Close modal

Although there was no difference in the level of [Ca2+]i mobilization between mitogen and CD3 activation, the response was stimulant concentration dependent. As shown in Fig. 2, A and B, lymphocytes from uninfected or infected mice responded differentially to varying concentrations of either anti-CD3 mAb or Con A. A significant difference was apparent between cells from infected vs uninfected mice.

FIGURE 2.

Effects of different concentrations of CD3 mAb (A) or Con A (B) stimulation on [Ca2+]i of lymphocytes obtained from either uninfected or day 5 postinfected mice. Lymphocytes from either uninfected or day 5 postinfection mice were loaded with indo-1 (45 min) and stimulated with different concentrations (5 and 1 μg/ml) of either anti-CD3 mAb or Con A. CD3 mAb or Con A was added 30 s before the beginning of the experiment, and the cells were analyzed at 250 cells/s. Results are plotted as the mean calcium concentration vs time.

FIGURE 2.

Effects of different concentrations of CD3 mAb (A) or Con A (B) stimulation on [Ca2+]i of lymphocytes obtained from either uninfected or day 5 postinfected mice. Lymphocytes from either uninfected or day 5 postinfection mice were loaded with indo-1 (45 min) and stimulated with different concentrations (5 and 1 μg/ml) of either anti-CD3 mAb or Con A. CD3 mAb or Con A was added 30 s before the beginning of the experiment, and the cells were analyzed at 250 cells/s. Results are plotted as the mean calcium concentration vs time.

Close modal

To better determine the specific T cell phenotype involved in the [Ca2+]i alteration postinfection, the [Ca2+]i level in CD4+ T cells from infected or uninfected mice was investigated. The CD4+ T cells were selected for study, since our data suggested that it was this population of T cells that was altered during acute infection (Table I). In these experiments, nylon wool-purified enriched T cells from both infected and uninfected mice were loaded with indo-1/AM. CD8+ T cells were removed by FACS following Ab staining. The residual cells to be used were approximately 90% positive for expression of CD4+ molecule. These cells were stimulated with either mitogen or anti-CD3 mAb, and the level of [Ca2+]i mobilization was determined. As illustrated in Fig. 3, CD4+ T cells obtained from infected mice showed a significant reduction in [Ca2+]i mobilization in response to stimulation via CD3 activation or mitogen. In response to anti-CD3 mAb exposure, the [Ca2+]i for these cells was reduced by one-third in the infected (320 nM) vs uninfected (480 nM) mice. Similarly, a 41% reduction was observed with Con A stimulation (uninfected, 550 nM; infected, 325 nM).

FIGURE 3.

Effects of CD3 mAb (A) or Con A (B) stimulation on [Ca2+]i of CD4+ lymphocytes. Indo-1-loaded T lymphocytes (from uninfected or day 5 postinfection mice) were stained for 20 min with FITC-conjugated CD8 mAb. The cells were analyzed at 250 cells/s, and electronic gating was used to display the FITC but CD4+ T cells. Different concentrations (5 and 1 μg/ml) of CD3 mAb or Con A were added 30 s before the beginning of the experiment, and the cells were analyzed at 250 cells/s. Results are plotted as the mean calcium concentration vs time.

FIGURE 3.

Effects of CD3 mAb (A) or Con A (B) stimulation on [Ca2+]i of CD4+ lymphocytes. Indo-1-loaded T lymphocytes (from uninfected or day 5 postinfection mice) were stained for 20 min with FITC-conjugated CD8 mAb. The cells were analyzed at 250 cells/s, and electronic gating was used to display the FITC but CD4+ T cells. Different concentrations (5 and 1 μg/ml) of CD3 mAb or Con A were added 30 s before the beginning of the experiment, and the cells were analyzed at 250 cells/s. Results are plotted as the mean calcium concentration vs time.

Close modal

We evaluated whether this alteration in calcium mobilization had an impact on the capacity to translocate NF-AT, an important transcription factor in activated T cells. Studies by others have shown, using an mAb (NF-ATc 7A6), that an elevation of intracellular calcium was required to maintain the NF-AT level in the nucleus. An immunofluorescence assay with this Ab was used to explore whether the reduced calcium flux in the lymphocytes from infected mice was sufficient for nuclear import of NF-AT (22, 29).

For this study mice were infected with T. gondii parasites, and their splenocytes were isolated on day 5 postinfection. The cells were stimulated with ionomycin and PMA, and the translocation of NF-AT was determined by fluorescence. When lymphocytes from uninfected mice were stimulated with ionomycin and PMA, NF-AT was imported into the nucleus (Fig. 4,B). In contrast, NF-AT could not be localized in the nucleus of stimulated T lymphocytes from acutely infected mice (Fig. 4,E). However, addition of a high concentration of CaCl2 (10 mM) resulted in translocation of NF-AT in the nuclei of lymphocytes from infected mice (Fig. 4 F).

FIGURE 4.

NF-ATc does not translocate in splenic cells obtained from day 5 postinfection animal. NF-ATc localization was assessed in splenocytes obtained from uninfected (A–C) and day 5 postinfection (D–F) animals by using NF-ATc mAb 7A6. Cells were cultured in medium alone for 60 min (A andD), stimulated with ionomycin (1 μM) and PMA (10 ng/ml) for 60 min (B and E), or stimulated with ionomycin (1 μM) and PMA (10 ng/ml) for 60 min but in the presence of 10 mM CaCl2 (C and F). Staining with isotype-matched control Ab gave no visible fluorescence.

FIGURE 4.

NF-ATc does not translocate in splenic cells obtained from day 5 postinfection animal. NF-ATc localization was assessed in splenocytes obtained from uninfected (A–C) and day 5 postinfection (D–F) animals by using NF-ATc mAb 7A6. Cells were cultured in medium alone for 60 min (A andD), stimulated with ionomycin (1 μM) and PMA (10 ng/ml) for 60 min (B and E), or stimulated with ionomycin (1 μM) and PMA (10 ng/ml) for 60 min but in the presence of 10 mM CaCl2 (C and F). Staining with isotype-matched control Ab gave no visible fluorescence.

Close modal

In this study we observed that acute murine infection with T. gondii results in generalized immunosuppression postinfection. During this period of immune unresponsiveness, splenocytes isolated from parasite-infected mice: 1) fail to proliferate in response to CD3 activation, 2) have diminished expression of both CD2 and CD4, 3) have an impaired [Ca2+]i response to mitogen stimulation, and 4) exhibit insufficient NF-AT import into the nucleus.

Diminished T cell proliferation during acute murine toxoplasmosis in response to cross-linked anti-CD3 mAb may be attributed to alterations in the mechanism of T cell activation. This condition was associated with an impairment in IL-2 production (6). The failure to restore T cell responsiveness by addition of exogenous IL-2 may be partly due to local factors defined by culture conditions, such as altered balances of cytokines and the inhibitory role of IL-10 and nitric oxide (4, 6, 7, 8, 30). CD48 augments the proliferative response of spleen cells when cross-linked with anti-CD3 mAbs (costimulatory signal) (31). In mice, CD48 is a ligand of CD2. We observed that cells from 5-day-postinfected mice were only partially responsive upon exposure to anti-CD48 mAb (data not shown).

The involvement of the CD4+ T cell subset during acute toxoplasma infection in mice has been investigated previously. Activation-induced programmed cell death may account for the unresponsive state of CD4+ T cells during acute infection (13). Alteration of the CD4/CD8 ratio during acute infection may preclude the progressive decline in the CD4+ population as the unresponsive T cells are sequestered and eliminated from the peripheral circulation by the process of apoptosis during in vitro culture. We evaluated this response in vivo using a chromatin condensation assay with Hoechst 33342. In that study we did not find any significant differences in the number of apoptic cells from either infected or uninfected mice (data not shown). The results of the current study demonstrated that splenocytes from infected mice exhibited decreased expression of CD4 on day 5 postinfection. A reduction in the expression of this surface molecule was observed in T cells from all three strains of mice investigated (CBA/J, BALB/c, and C57BL/6). Several reports suggest that the CD4 molecule may serve to enhance or inhibit CD3-induced signaling, although we observed no changes in the expression of CD3 (32). Although the modulation of CD4+ molecule was identical in the different strains of mice used in our experiments, alterations in CD2 were most apparent in the highly susceptible C57 mouse strain. The contribution of CD2 to the immune hyporesponsiveness in vivo has been difficult to assess due to the limitation of experimentation in animal models (33, 34, 35). Studies in HIV-infected cells demonstrate the persistence of CD2 mRNA expression despite a marked diminution in its surface expression (18). The importance of CD2 regulation in the development of immune unresponsiveness to toxoplasma infection remains uncertain and is currently under study.

In some parasitic protozoan infections such as T. cruzi (21, 36, 37), Plasmodium falciparum (38), and Leishmania donovani (19) infections, a role for Ca+ in the process of host cell invasion has been suggested. These studies indicated changes in the cytosolic Ca+ concentration in diverse host cells, such as in HUVEC, fibroblast cells, HeLa cells, and mononuclear phagocytes, following infections by these parasites. However, little information is available on the consequences of these changes on the host’s immune responses. Few studies have provided information on calcium signal transduction in T cells during the course of parasitic infections. In the present study we report that during acute toxoplasma infection lymphocytes obtained from infected mice had a significant reduction in Ca+ mobilization (50–53% reduction in the [Ca+]i response to both CD3 mAb and mitogen compared with control values) (Fig. 1). The importance of Ca2+ in T lymphocyte activation is evident from the effectiveness of the immunosuppressant cyclosporin A and the observations that individuals with lymphocytes defective in Ca2+ signaling suffer from primary immunodeficiency (39). HIV has been shown to alter the stores of intracellular free calcium and impair inositol phosphatase production (18). Our findings clearly demonstrate that T. gondii, like HIV, can affect stores of intracellular calcium and suggest that a reduced calcium flux could be implicated in the development of transient T cell hyporesponsiveness during acute infection.

We next wanted to determine whether the Ca+ signaling defect in stimulated T cells from infected mice could regulate NF-AT, the transcription factor that stimulates early immune response genes such as cytokines (23, 40). A sustained rise in the intracellular Ca2+ concentration can activate calcineurin, a Ca2+-dependent, cyclosporin A-sensitive serine/threonine phosphatase that dephosphorylates the transcription factor NF-AT (23). Once dephosphorylated, NF-AT migrates to the nucleus, where it associates with Jun and Fos to promote the transcription of a host of immunoregulatory genes (40). We have taken advantage of availability of a mAb to NF-ATc (7A6) (25), which is expressed exclusively in the lymphoid system and is induced upon lymphocyte activation. By using this mAb in confocal microscopy, we were able to evaluate calcium regulation of NF-AT translocation in the nucleus of T lymphocytes stimulated with ionomycin and PMA. Our data clearly showed that the calcium flux in the lymphocytes from infected mice was not sufficient for nuclear import of NF-AT (compare Fig. 4, D andE with A and B). However, NF-AT will translocate in the nucleus of these lymphocytes if [Ca2+]i is artificially augmented and sustained by increasing concentrations of extracellular Ca2+ (Fig. 4 F). Ca+ signaling involves the mobilization of Ca+ from intracellular stores and the extracellular medium (41). We and others have demonstrated that a decrease in both the production and the expression of IL-2 occurs during the hyporesponsive state associated with acute toxoplasma infection (6, 8, 42). Insufficient concentrations of intracellular free calcium may explain the defect in IL-2 production by T lymphocytes from acutely infected mice. It may be noted that sustained levels of Ca+ are required to maintain NF-AT transcription factors in the nucleus, where they participate in Ca+-dependent induction of genes required for IL-2 enhancement (23, 40). Together these findings indicate that during acute toxoplasma infection, NF-AT translocation is affected inside T lymphocytes by live parasite infection, rendering the host unable to respond in an immunologically competent fashion.

We thank Dr. Luika Timmerman (Stanford University School of Medicine, Palo Alto, CA) for her generous gift of the valuable reagents (Abs of the NF-AT family) without which immunofluorescence for NF-AT family member visualization would not have been possible. We are grateful to Drs. Michael W. Fagner (Dartmouth Medical School, Hanover, NH) and Christopher Fagner (Department of Molecular and Cellular Physiology, Stanford University) for their suggestions and help. We thank Dr. Jacquline Channon for critical reading of the manuscript, Drs. Claude Boyer (Centre d’Immunologie Luminy, Marseille, France) and Alice Given (Dartmouth Medical School, Hanover, NH) for helpful discussion and technical advice during the preparation of Ca2+ assay, and Gary Ward and Kenneth A. Orndorf (Dartmouth Medical School, Hanover, NH) for their expert help with flow cytometric analysis and photoshop.

1

This work was supported by Grants AI35956 and AI30000 from the National Institutes of Health.

3

Abbreviations used in this paper: [Ca2+]i, intracellular Ca2+; PE, phycoerythrin; LP, live tachyzoite of Toxoplasma gondii parasite; FFP, formalin-fixed tachyzoite of Toxoplasma gondii parasite.

1
Anderson, S. E., J. L. Krahenbuhl, Jr, J. S. Remington.
1979
. Longitudinal studies of lymphocyte response to toxoplasma antigens in humans infected with T.
gondii. J. Clin. Lab. Immunol.
2
:
293
2
Luft, B. J., G. Kansas, E. G. Engleman, J. S. Remington.
1984
. Functional and quantitative alterations in T lymphocyte subpopulations in acute toxoplasmosis.
J. Infect. Dis.
150
:
761
3
Sher, A., I. P. Oswald, S. Hieny, R. T. Gazzinelli.
1993
. Toxoplasma gondii induces a T-independent IFN-γ response in natural killer cells that requires both adherent accessory cells and tumor necrosis factor-α.
J. Immunol.
150
:
3982
4
Channon, J. Y., L. H. Kasper.
1996
.
Toxoplasma gondii-induced immune suppression by human peripheral blood monocytes: role of γ interferon. Infect. Immun.
64
:
1181
5
Candolfi, E., C. A. Hunter, J. S. Remington.
1994
. Mitogen- and antigen-specific proliferation of T cells in murine toxoplasmosis is inhibited by reactive nitrogen intermediates.
Infect. Immun.
62
:
1995
6
Haque, S., I. Khan, A. Haque, L. H. Kasper.
1994
. Impairment of the cellular immune response in acute murine toxoplasmosis: regulation of interleukin 2 production and macrophage-mediated inhibitory effects.
Infect. Immun.
62
:
2908
7
Suzuki, Y., K. Joh, A. Kobayashi.
1987
. Macrophage-mediated suppression of immune responses in Toxoplasma-infected mice. III. Suppression of antibody responses to parasite itself.
Cell. Immunol.
110
:
218
8
Haque, S., A. Haque, L. H. Kasper.
1995
. A Toxoplasma gondii-derived factor(s) stimulates immune downregulation: an in vitro model.
Infect. Immun.
63
:
3442
9
Denkers, E. Y., R. T. Gazzinelli, D. Martin, A. Sher.
1993
. Emergence of NK1.1+ cells as effectors of IFN-γ dependent immunity to Toxoplasma gondii in MHC class I-deficient mice.
J. Exp. Med.
5
:
1465
10
Hunter, C. A., R. Chizzonite, J. S. Remington.
1995
. IL-1 beta is required for IL-12 to induce production of IFN-γ by NK cells: a role for IL-1β in the T cell-independent mechanism of resistance against intracellular pathogens.
J. Immunol.
155
:
4347
11
Khan, I. A., K. H. Ely, L. H. Kasper.
1994
. An antigen specific CD8+ T cell clone protects against acute T.
gondii infection in mice. J. Immunol.
152
:
1856
12
Denkers, E. Y., P. Caspar, S. Hieny, A. Sher.
1996
. Toxoplasma gondii infection induces specific nonresponsiveness in lymphocytes bearing the Vβ5 chain of the mouse T cell receptor.
J. Immunol.
156
:
1089
13
Khan, I. A., T. Matsuura, L. H. Kasper.
1996
. Activation-mediated CD4+ T cell unresponsiveness during acute Toxoplasma gondii infection in mice.
Int. Immunol.
8
:
887
14
Hara, T., L. K. Jung, J. M. Bjorndahl, S. M. Fu.
1986
. Human T cell activation. III. Rapid induction of a phosphorylated 28 kD/32 kD disulfide-linked early activation antigen (EA 1) by 12-O-tetradecanoyl phorbol-13-acetate, mitogens, and antigens.
J. Exp. Med.
164
:
1988
15
Testi, R., J. H. Phillips, L. L. Lanier.
1989
. Leu 23 induction as an early marker of functional CD3/T cell antigen receptor triggering: requirement for receptor cross-linking, prolonged elevation of intracellular [Ca++] and stimulation of protein kinase C.
J. Immunol.
142
:
1854
16
Harding, F. A., J. G. McArthur, J. A. Gross, D. H. Raulet, J. P. Allison.
1992
. CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones.
Nature
356
:
607
17
Van Wauwe, J., J. Goossens, W. Decock, P. Kung, G. Goldstein.
1981
. Suppression of human T-cell mitogenesis and E-rosette formation by the monoclonal antibody OKT11A.
Immunology
44
:
865
18
Linette, G. P., R. J. Hartzman, J. A. Ledbetter, C. H. June.
1988
. HIV-1-infected T cells show a selective signaling defect after perturbation of CD3/antigen receptor.
Science
241
:
573
19
Olivier, M., K. G. Baimbridge, N. E. Reiner.
1992
. Stimulus-response coupling in monocytes infected with Leishmania: attenuation of calcium transients is related to defective agonist-induced accumulation of inositol phosphates.
J. Immunol.
148
:
1188
20
Turco, S. J., A. Descoteaux.
1992
. The lipophosphoglycan of Leishmania parasites.
Annu. Rev. Microbiol.
46
:
65
21
Morris, S. A., H. Tanowitz, V. Hatcher, J. P. Bilezikian, M. Wittner.
1988
. Alterations in intracellular calcium following infection of human endothelial cells with Trypanosoma cruzi.
Mol. Biochem. Parasit.
29
:
213
22
Fanger, C. M., M. Hoth, G. R. Crabtree, R. S. Lewis.
1995
. Characterization of T cell mutants with defects in capacitative calcium entry: genetic evidence for the physiological roles of CRAC channels.
J. Cell Biol.
131
:
655
23
Crabtree, G. R., N. A. Clipstone.
1994
. Signal transmission between the plasma membrane and nucleus of T lymphocytes.
Annu. Rev. Biochem.
63
:
1045
24
Dolmetsch, R. E., R. S. Lewis, C. C. Goodnow, J. I. Healy.
1997
. Differential activation of transcription factors induced by Ca2+ response amplitude and duration.
Nature
386
:
855
25
Timmerman, L. A., N. A. Clipstone, S. N. Ho, J. P. Northrop, G. R. Crabtree.
1996
. Rapid shuttling of NF-AT in discrimination of Ca2+ signals and immunosuppression.
Nature
383
:
837
26
Lucas, B., L. H. Kasper, K. Smith, A. Haque.
1996
. In vivo treatment with interleukin 2 reduces parasitemia and restores IFN-γ gene expression and T-cell proliferation during acute murine malaria.
C. R. Acad. Sci. Ser. Iii Life Sci.
319
:
705
27
Rabinovitch, P. S., C. H. June, A. Grossmann, J. A. Ledbetter.
1986
. Heterogeneity among T cells in intracellular free calcium responses after mitogen stimulation with PHA or anti-CD3: simultaneous use of indo-1 and immunofluorescence with flow cytometry.
J. Immunol.
137
:
952
28
Grynkiewicz, G., M. Poenie, R. Y. Tsien.
1985
. A new generation of Ca2+ indicators with greatly improved fluorescence properties.
J. Biol. Chem.
260
:
3440
29
Serafini, A. T., R. S. Lewis, N. A. Clipstone, R. J Bram, C. Fanger, S. Fiering, L. A. Herzenberg, G. R. Crabtree.
1995
. Isolation of mutant T lymphocytes with defects in capacitative calcium entry.
Immunity
3
:
239
30
Khan, I. A., T. Matsuura, L. H. Kasper.
1995
. IL-10 mediates immunosuppression following primary infection with Toxoplasma gondii in mice.
Parasit. Immunol.
17
:
185
31
Kato, K., M. Koyanagi, H. Okada, T. Takanashi, Y. W. Wong, A. F. Williams, K. Okumura, H. Yagita.
1992
. CD48 is a counter receptor for mouse CD2 and is involved in T cell activation.
J. Exp. Med.
176
:
1241
32
Tite, J. P., A. Sloan, C. A. Janeway.
1986
. The role of L3T4 in T cell activation: L3T4 may be both an Ia-binding protein and a receptor that transduces a negative signal.
J. Mol. Cell. Immunol.
2
:
179
33
Clark, S. J., D. A. Law, D. J. Paterson, M. Puklavec, A. F. Williams.
1988
. Activation of rat T lymphocytes by anti-CD2 monoclonal antibodies.
J. Exp. Med.
167
:
1861
34
Guckel, B., C. Berek, M. Lutz, P. Altevogt, V. Schirrmacher, B. A. Kyewski.
1991
. Anti-CD2 antibodies induce T cell unresponsiveness in vivo.
J. Exp. Med.
174
:
957
35
Meuer, S. C., R. E. Hussey, M. Fabbi, D. Fox, O. Acuto, K. A. Fitzgerald, J. C. Hodgdon, J. P. Protentis, S. F. Schlossman, E. L. Reinherz.
1984
. An alternative pathway of T-cell activation: a functional role for the 50 kd T11 sheep erythrocyte receptor protein.
Cell
36
:
897
36
Low, H. P., J. J. Paulin, C. H. Keith.
1992
. Trypanosoma cruzi infection of BSC-1 fibroblast cells causes cytoskeletal disruption and changes in intracellular calcium levels.
J. Protozool.
39
:
463
37
Osuna, A., S. Castanys, M. N. Rodriguez-Cabezas, F. Gamarro.
1990
.
Trypanosoma cruzi: calcium ion movement during internalization in host HeLa cells. Int. J. Parasitol.
20
:
673
38
Adovelande, J., B. Bastide, J. Deleze, J. Schrevel.
1993
. Cytosolic free calcium in Plasmodium falciparum-infected erythrocytes and the effect of verapamil: a cytofluorometric study.
Exp. Parasitol.
76
:
247
39
Partiseti, M., F. Le Deist, C. Hivroz, A. Fischer, H. Korn, D. Choquet.
1994
. The calcium current activated by T cell receptor and store depletion in human lymphocytes is absent in a primary immunodeficiency.
J. Biol. Chem.
269
:
32327
40
Rao, A..
1994
. NF-ATp: a transcription factor required for the co-ordinate induction of several cytokine genes.
Immunol. Today
15
:
274
41
Docampo, R., S. N. J. Moreno.
1996
. Role of Ca+ in the process of cell invasion by intracellular parasites.
Parasitol. Today
12
:
61
42
Chan, J., J. P. Siegel, B. J. Luft.
1986
. Demonstration of T-cell dysfunction during acute toxoplasma infection.
Cell. Immunol.
98
:
422