Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Anti-CD25 Antibody-Mediated Depletion of Effector T Cell Populations Enhances Susceptibility of Mice to Acute but Not Chronic Toxoplasma gondii Infection

Kevin N. Couper, Paula A. Lanthier, Georgia Perona-Wright, Lawrence W. Kummer, Wangxue Chen, Stephen T. Smiley, Markus Mohrs and Lawrence L. Johnson
J Immunol April 1, 2009, 182 (7) 3985-3994; DOI: https://doi.org/10.4049/jimmunol.0803053
Kevin N. Couper
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Paula A. Lanthier
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Georgia Perona-Wright
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lawrence W. Kummer
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wangxue Chen
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stephen T. Smiley
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Markus Mohrs
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lawrence L. Johnson
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Natural regulatory T cells (Tregs) constitutively express the IL-2R α-chain (CD25) on their surface. Consequently, administration of anti-CD25 Abs is a commonly used technique to deplete Treg populations in vivo. However, activated effector T cells may also transiently express CD25, and are thus also potential targets for anti-CD25 Abs. In this study using Toxoplasma gondii as a model proinflammatory infection, we have examined the capacity of anti-CD25 Abs to target effector T cell populations during an inflammatory episode, to determine to what extent that this action may modulate the outcome of disease. Anti-CD25 Ab-treated C57BL/6 mice displayed significantly reduced CD4+ T cell IFN-γ production during acute T. gondii infection and exhibited reduced weight loss and liver pathology during early acute infection; aspects of infection previously associated with effector CD4+ T cell responses. In agreement, anti-CD25 Ab administration impaired parasite control and caused mice to succumb to infection during late acute/early chronic stages of infection with elevated tissue parasite burdens. In contrast, anti-CD25 Ab treatment of mice with established chronic infections did not markedly affect brain parasite burdens, suggesting that protective T cell populations do not express CD25 during chronic stages of T. gondii infection. In summary, we have demonstrated that anti-CD25 Abs may directly abrogate effector T cell responses during an inflammatory episode, highlighting important limitations of the use of anti-CD25 Ab administration to examine Treg function during inflammatory settings.

Natural regulatory T cells (Tregs)3 constitutively express high levels of the IL-2R α-chain (CD25) and until recently this marker was the most commonly used to identify these cells. Natural Tregs, which comprise 5–10% of CD4+ T cells in naive mice, have potent regulatory capacity and have been shown to have important roles in a number of autoimmune and infectious diseases (reviewed in Refs. 1 , 2). Importantly, however, CD25 is not a specific marker of natural Tregs and it may be expressed by a number of other cell populations, including activated T and B cells, monocytes, and some dendritic cells (reviewed in Ref. 3). Moreover, the identification of the transcription factor Foxp3 as a lineage specific marker for natural Tregs has enabled more accurate differentiation of regulatory and effector CD4+ T cell populations, and has shown that populations of CD25− Foxp3+ Tregs also exist (4).

Nevertheless, administration of anti-CD25 Abs remains a commonly used method to deplete natural Tregs in vivo. Some studies have obtained results consistent with the depletion of regulatory cell populations, as evidenced by augmented protective immune responses and enhanced pathogen control (for example see Refs. 5, 6, 7, 8), whereas other studies have observed no difference in immunity following anti-CD25 administration, possibly due to the disease model used or the strain and immune status of mice in the study (for example see Refs. 9, 10, 11, 12, 13). In addition, anti-CD25 Abs are also routinely used in vitro for the positive selection of natural Tregs from naive and infected mice for the subsequent use in adoptive transfer systems or in vitro suppression assays. In general, these experiments have almost entirely focused on the effects of anti-CD25 Abs on CD4+ natural Tregs, even though many cells other than natural Tregs, including effector T cells, could potentially be targeted by anti-CD25 treatment (3). Thus, anti-CD25 treatment in vivo may have effects beyond simply depleting CD25-expressing natural Tregs or affecting their regulatory capability, potentially leading to inaccurate conclusions on the role of natural Tregs in any particular disease setting.

In this study, we use oral Toxoplasma gondii infection as a model of inflammatory disease to directly investigate the effect of anti-CD25 Ab treatment on the effector arm of the immune response. The T. gondii infection model has several useful features for this purpose. During acute infection, which lasts 1–2 wk, strong proinflammatory innate and adaptive responses develop as rapidly proliferating single cell tachyzoites develop from dormant encysted parasites and disseminate from the intestine to liver, lung, brain, and other sites. Cooperation among neutrophils, NK cells, and macrophages is required for the production of IL-12 and IFN-γ, which then play critical protective roles during both acute and chronic infections (14, 15, 16, 17, 18, 19). However, in susceptible mouse strains such as C57BL/6 (B6), type I cytokines cause immunopathology in small intestine and liver that is not evident in resistant mouse strains (e.g., BALB/c) (20). Notably, CD4+ cells, although not required for control of parasites in the first week or so of acute infection, are nonetheless important mediators of the immunopathology seen in C57BL/6 mice (20).

Following the development of sufficient adaptive T cell-mediated immune responses, the acute stage of infection is controlled, and tachyzoites are cleared in the intestine, liver, and lungs and parasites revert to a semidormant state within cysts in the brain. In this chronic infection phase, acquired immune mechanisms dependent on CD4+ and CD8+ T cells and B cells, along with IFN-γ and TNF-α are required to control tachyzoite dissemination and to prevent the development of toxoplasmic encephalitis (21, 22, 23, 24). Thus, activated effector CD4+ T cells mediate damaging immunopathology early during acute infection, yet contribute to parasite control during late acute/chronic stages of infection and prevent the development of encephalitis during chronic infection.

Our results support the conclusion that anti-CD25 Ab administration directly targets CD25 expressing effector CD4+ T cells during acute T. gondii infection. We show that anti-CD25 Ab administration, while depleting ∼40% of Foxp3+ natural Tregs, also significantly reduces effector CD4+ T cell numbers and diminishes IFN-γ production during acute T. gondii infection, ameliorating early immune-mediated pathology but significantly impairing disease control and elevating mortality during late acute/early chronic stage T. gondii infection. Thus, our results strongly imply that anti-CD25 Abs should be used with caution in vivo during highly proinflammatory disease models, and that the potential effects on all cells that can express CD25 should be considered when they are used to examine the function and importance of natural Tregs.

Materials and Methods

Mice

C57BL/6J, Thy1.1 C57BL/6 (B6), and bicistronic IFN-γ reporter (Yeti) male mice (25) were used between 6 and 12 wk of age. Mice were bred in the Trudeau Institute Animal Breeding Facility or obtained from The Jackson Laboratory or Taconic Farms. All experiments were reviewed and approved by the Trudeau Institute Institutional Animal Care and Use Committee. The Trudeau Institute is fully accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care.

Parasites and infections

ME49 cysts were obtained from brains of chronically infected B6 mice. Infections were initiated by peroral administration of 10 cysts in 0.1 ml of diluted brain suspension as previously described (26). Sham-infected mice received similarly diluted brain suspension containing no cysts. Weight loss was monitored as a sign of morbidity and is expressed as the percentage of baseline starting weight. Mice were monitored daily for survival.

Ab treatments

Mice were given i.p. injections of 1 mg of anti-CD25 (clone PC61) or a control rat IgG (clone HRPN) obtained from BioXcell (previously BioExpress). Abs were administered 5–7 days before infection or during infection on days stated in the experiment. PC61 administration promoted long-lasting depletion/neutralization of CD25 expression (longer than 2 wk) in naive mice, consistent with previously published reports (27). Endotoxin levels of clones PC61 and HRPN were below 1 EU/mg.

Flow cytometry

Single cell suspensions were prepared from spleens, liver, blood, and mesenteric lymph nodes. Brains were gently mashed using a Teflon pestle, cell populations separated on a Percoll gradient, and lymphocytes recovered for analysis. Cells were counted using a hemacytometer. The surface Ag phenotype was determined by flow cytometry using the following fluorochrome-conjugated Abs: anti-CD4 (clone GK1.5), anti-CD25 (clone PC61 or clone 7D4), or anti-CD44 (clone IM7). Cells were preincubated with Fc block (clone 2.4G2) before staining. All Abs were obtained from BD Biosciences. Foxp3 staining was performed using anti-Foxp3 (FJK-16s) Ab according to the manufacturer’s instructions (eBioscience). It is important to note that fluorochrome-labeled 7D4 was used for flow cytometric detection of CD25 expression in all experiments in which PC61 was administered in vivo to deplete CD25-expressing cells; PC61 does not inhibit 7D4 detection of cell surface CD25 (27).

Isolation of CD4+CD25− and CD4+CD25high T cells

CD4+ T cells were purified from the spleens and brains of C57BL/6 mice using anti-CD4 MidiMacs beads according to the manufacturer’s instructions (Miltenyi Biotec). Following separation, CD4+ cells were stained with anti-CD4 (GK1.5) and anti-CD25 (PC61) and sorted into CD4+CD25− and CD4+CD25high populations using a FACSVantage cell sorter. The sorted cell populations were routinely >99% pure.

In vitro restimulation experiments

CD4+CD25high and CD4+CD25− cells, obtained as described, were cultured individually or at a ratio of 1:1. Cells were either stimulated with naive APCs and anti-CD3 Ab (1 μg/ml) or with T. gondii-pulsed APC (without anti-CD3 Ab costimulation). APCs were prepared by gamma irradiation (3000 rad) of spleen cells obtained from naive B6 mice. Separately, T. gondii primed APC were prepared to measure parasite-specific T cell responses. RH strain tachyzoites, maintained through continuous in vitro culture in fibroblast monolayers, were irradiated at 15,000 rad and added to APC generated from naive mice as indicated. Irradiated tachyzoites and APC were cultured for 12 h at 5% CO2 and 37°C before being washed and added at required cell concentration to wells including purified CD4+ cells. The cells were then incubated at 5% CO2 and 37°C for 60 h. Supernatants were then harvested to assay cytokine production and proliferation was determined by pulsing cells with 0.2 μCu/well [3H]thymidine (Amersham Biosciences) for an extra 12 h. [3H]Thymidine incorporation and proliferation was measured using a beta counter.

ELISA and real-time PCR

Serum IFN-γ levels were measured by ELISA as previously described (18). Tachyzoites were quantified by a real-time PCR-based method (28) based on the expression of the tachyzoite-limited Sag1 gene (29). RNA was isolated from tissues, cDNA prepared, amplified, and quantitated using the following primer to probe sets: T. gondii Sag1 (forward) TTTCCGAAGGCAGTGAGACG, (reverse) GGATCCGATGCCATAGCG, probe TTGCCGCGCCCACACTGATG; IFN-γ (forward) CATTGAAAGCCTAGAAAGTCTGAATAAC, (reverse) TGGCTCTGCAGGATTTTCATG, probe TCACCATCCTTTTGCCAGTTCCTCCAG; IL-10 (forward) GAAGACCCTCAGGATGCGG, (reverse) ACCTGCTCCACTGCCTTGCT, probe TGAGGCGCTGTCATCGATTTCTCCC; Foxp3 (forward) CCCAGGAAAGACAGCAACCTT, (reverse) TTCTCACAACCAGGCCACTTG, probe ATCCTACCCACTGCTGGCAAATGGAGTC; and GAPDH (forward) CTCGTCCCGTAGACAAAATGG, (reverse) AATCTCCACTTTGCCACTGCA, probe CGGATTTGGCCGTATTGGGCG. Values were normalized to GAPDH expression. IFN-γ message levels in brain are shown as fold change (log10) relative to control mAb-treated infected mice. Tachyzoite numbers were calculated relative to a standard curve generated by inoculating known numbers of tachyzoites into uninfected liver samples.

Histological exam

On day 8 postinfection, livers and ileums from anti-CD25 Ab-treated mice and control Ab-treated mice were removed and processed for H&E staining. Liver and ileal lesions were semiquantitatively scored based on our previous scoring systems (28, 30) as follows: liver damage 1, degeneration and necrosis of individual hepatocytes occasionally seen; 2, clusters or small aggregates of hepatocyte degeneration and necrosis; 3, medium to large aggregates of hepatocyte necrosis; 4, large areas of hepatocyte necrosis with loss of normal liver anatomic architecture; intensity of the inflammatory response: 1, small inflammatory infiltrates with a few inflammatory cells; 2, medium-sized inflammatory infiltrates with a small to moderate number of inflammatory cells; 3, large inflammatory infiltrates with a moderate to large number of inflammatory cells; 4, extensive infiltration with a large number of inflammatory cells. The ileal lesions were scored for the degeneration and necrosis of villous epithelial cells, transmural inflammation, and lamina propria and crypt inflammation and an integer score between 0 (unremarkable) and 4 (severe) was assigned.

Statistics

Unless otherwise noted, group means were compared by Student’s t test. Intergroup survivals were compared by log-rank test. All statistical analyses were performed using GraphPad Prism software.

Results

Anti-CD25 Ab increases susceptibility of mice to late acute T. gondii infection

The administration of anti-CD25 Ab (clone PC61) significantly increased the susceptibility of mice to acute oral T. gondii infection (Fig. 1⇓). The majority of C57BL/6 mice treated with PC61 succumbed to infection by day 20 of infection, and this correlated with a biphasic modulation of weight loss (Fig. 1⇓, A and B). PC61 pretreated mice exhibited reduced weight loss during early acute T. gondii infection, with significant differences compared with control Ab treated mice on days 6–8 of infection (p < 0.01). However, in contrast to control Ab-treated mice that survived acute infection and recovered weight, PC61-treated mice were unable to resolve the acute infection and exhibited significantly elevated weight loss during mid to late acute infection on days 15–18 (p < 0.05) (Fig. 1⇓A). These data are consistent with previous studies in which the depletion of proinflammatory CD4+ T cells ameliorated weight loss and pathology during early acute infection but caused enhanced weight loss during late acute infection (31). Surviving PC61 pretreated mice also harbored significantly increased brain cyst numbers compared with control mice on day 30 postinfection (p < 0.033) (Fig. 1⇓C).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Anti-CD25 Ab administration significantly modulates the outcome of acute T. gondii infection. Groups of five B6 males were given 1 mg of control mAb (clone HRPN) or anti-CD25 (clone PC61) 5 or 7 days before oral infection with 10 ME49 cysts. A, Changes in body weight were monitored and are presented as a percentage change relative to starting weight. Data show mean ± SD. ∗, p < 0.05 for significant difference. B, Compiled survival data from three experiments comparing anti-CD25-treated with control T. gondii-infected B6 male mice. Survivals differed significantly (p < 0.01) by log-rank test. C, Brain cysts of three anti-CD25-treated B6 male mice, and five controls 28 days after mice were sham-infected or infected with 10 ME49 cysts. Horizontal bar denotes median values. D, On day 8 postinfection, livers and ileums were removed and stained with H & E for histological examination of tissue pathology with arrows marking areas of inflammation or necrosis (also marked by an asterisk). E and F, On day 8 postinfection, livers (E) and ileums (F) were removed for real-time PCR (Taqman) quantification of tissue parasite burdens. Results shown are mean ± SD of the group (n = 5) and are representative of two independent experiments. ∗∗, p < 0.01.

Anti-CD25 Abs modulate development of immune-mediated pathology and significantly impair parasite control during acute T. gondii infection

As anti-CD25 Ab administration elevated mortality, modulated weight loss, and increased brain cyst numbers, we next examined whether anti-CD25 Ab administration impeded tachyzoite control or limited acute stage immunopathology. In agreement with this hypothesis, we observed significantly reduced liver pathology in anti-CD25 Ab-treated mice compared with control Ab-treated mice on day 8 of infection, with ameliorated inflammatory cell infiltration (predominantly mononuclear cells and neutrophils) throughout the parenchymal and portal areas and significantly reduced levels of hepatic coagulation necrosis (Fig. 1⇑D and Table I⇓). In addition, moderately severe perivasculitis, vasculitis, and fibrinous thrombosis was observed in medium to large blood vessels of control Ab-treated mice, which was relatively absent in anti-CD25 Ab-treated mice (Fig. 1⇑D). Nevertheless, parasite control in the liver was relatively unaffected by anti-CD25 Ab treatment, as comparable tachyzoite numbers were observed in control Ab- and PC61-treated mice on day 8 postinfection (Fig. 1⇑E), indicating that parasite control in the liver during early acute T. gondii infection is CD25-independent. In contrast, significantly elevated pathology was observed in the ileum of anti-CD25 Ab-treated mice compared with control Ab-treated mice on day 8 postinfection; extensive infiltration of a moderate number of mononuclear cells was observed in the lamina propria of anti-CD25 Ab-treated mice, which was absent in control Ab-treated mice (Fig. 1⇑D and Table I⇓). Elevated pathology was directly correlated with increased parasite burdens in the ileum of anti-CD25 Ab-treated mice on day 8 postinfection (Fig. 1⇑F), suggesting that the failure of parasite control, rather than partial depletion or inactivation of natural Tregs following anti-CD25 Ab administration, was responsible for the increased pathology evident in the ileum. Combined, these data highlight the pathological role of CD25-expressing effector cells in mediating hepatic pathology during early acute T. gondii infection and demonstrate the importance of CD25-expressing effector cells for control of T. gondii parasites in the ileum during the acute stage of infection.

View this table:
  • View inline
  • View popup
Table I.

Tissue pathology during acute T. gondii infection following anti-CD25 Ab administration

CD25 is transiently up-regulated on CD4 T cells during acute T. gondii infection

To examine the kinetics of CD25 expression during T. gondii infection, to establish when it is up-regulated and on which cells, we determined the expression level of CD25 on CD4+ and CD8+ splenic lymphocytes on various days during acute T. gondii infection. Fig. 2⇓A shows the marked increase between days 4 and 12 of infection in both the absolute number of total splenic CD4+ T cells and the number of splenic CD4+ T cells expressing high levels of CD25. A similar transient up-regulation of CD25 on splenic CD8+ T cells was also observed between days 4 and 12 of infection, before absolute numbers of splenic CD8+ T cells and CD8+CD25high T cells returned to baseline preinfection levels (results not shown).

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Transient up-regulation of CD25 on effector T cells during acute T. gondii infection. A, Total number of splenic CD4+ T cell and CD4+CD25high T cells was calculated on selected days following oral T. gondii infection. CD25 detection was performed using the clone PC61 anti-CD25 Ab. Error bar shows mean ± SD. Number above bar for CD4+CD25high cells indicates the percentage of that population within the total CD4+ T cell population. B, Cell sorted Thy1.1+CD4+CD25− cells were adoptively transferred into Thy1.2+ B6 mice before oral T. gondii infection. The frequency of adoptively transferred donor cells expressing CD25 in the spleen was determined on selected days postinfection. C and D, In a separate experiment, groups of five B6 males were infected as in A, and spleens harvested on day 8 to analyze the percentage of CD4+CD25high cells that coexpressed Foxp3. The number of CD4+CD25high cells and CD4+Foxp3+ cells (C) and representative dot plots (D) gating on CD4+ T cells showing CD25 expression vs Foxp3 expression in naive and T. gondii-infected mice. Mean + SD is indicated for CD4+ populations. ∗, p < 0.01.

The expansion of CD4+ T cells expressing CD25 during acute T. gondii infection could potentially have been due to either the proliferation of CD4+CD25high Tregs (present in naive mice) or due to the up-regulation of CD25 on activated precursor CD25− naive CD4+ T cells. Thus, to investigate the ability of CD4+CD25− T cells to up-regulate CD25 during acute T. gondii infection we purified and adoptively transferred Thy1.1+CD4+CD25− cells into congenic Thy1.2+ mice before oral T. gondii infection. Importantly, significant up-regulation of CD25 expression was observed on the CD4+CD25− donor T cell population (Fig. 2⇑B). Moreover, the kinetics (and magnitude) of CD25 up-regulation on the donor CD4+ T cells mirrored the up-regulation of CD25 in the total CD4+ T cell population in wild-type mice (Fig. 2⇑A), indicating that CD4+CD25− cells are the primary precursor cells of the CD4+CD25high cells generated during acute T. gondii infection.

Although there were significantly more CD4+CD25high spleen cells in infected mice than in sham-infected mice on day 8 postinfection, there was no significant difference in the number of total CD4+Foxp3+ cells, or the number of CD4+Foxp3+ cells expressing CD25 (Fig. 2⇑C), suggesting that the majority of CD4+CD25high T cells were not Tregs, as shown from day 8 of infection (Fig. 2⇑D). Moreover, a significant number of CD4+Foxp3+ cells were found within the CD25low subset of CD4+ cells, revealing the presence of Foxp3+CD25− Tregs during T. gondii infection and in naive uninfected mice (Fig. 2⇑C).

CD4+CD25high T cells are primarily effector T cells during acute T. gondii infection

It is well established that CD4+CD25high T cells derived from naive mice express high levels of Foxp3 and exhibit potent regulatory capabilities. In contrast, extremely strong systemic proinflammatory immune responses develop during T. gondii infection and the majority of CD4+CD25high cells do not express Foxp3 (Fig. 2⇑), indicating that these cells may exert effector rather than regulatory functions. To address this likelihood, we purified CD4+CD25− and CD4+CD25high cells from uninfected mice and mice acutely infected with T. gondii and assessed the ability of these cells to respond to mitogenic (anti-CD3) and T. gondii Ag-specific stimulation. As expected, CD4+CD25high cells derived from uninfected mice closely matched the functional characteristics of natural Tregs; they failed to produce IFN-γ or proliferate following mitogenic stimulation and suppressed proliferation and IFN-γ production by CD4+CD25− cells (p < 0.05) (Fig. 3⇓, A and B). Conversely, CD4+CD25high cells derived from acutely infected mice failed to suppress CD4+CD25− cell responses, produced significant quantities of IFN-γ and proliferated rapidly following mitogenic and T. gondii antigenic challenge (Fig. 3⇓, C–F). Importantly, CD4+CD25high T cells derived from acutely infected mice produced more IFN-γ and proliferated more rapidly than infection-derived CD4+CD25− cells following specific T. gondii stimulation (Fig. 3⇓, E and F), suggesting that during acute T. gondii infection, the majority of parasite-specific effector CD4+ T cells are found within the CD4+CD25high T cell population.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

CD4+ T cells that up-regulate CD25 expression during acute T. gondii infection are Th1 effector cells that produce high levels of IFN-γ. CD4+CD25− and CD4+CD25high splenic T cells were sort purified from naive mice (A and B) or T. gondii-infected mice (B–F) (day 10 of infection). CD25 detection was performed using the PC61 clone anti-CD25 Ab. CD4+CD25− and CD4+CD25high cells were cultured with naive APC and anti-CD3 Ab (A–D) or T. gondii-pulsed APC (E and F). A, C, and E, IFN-γ production was assayed in supernatant by ELISA. B, D, and F, Proliferation was determined by [3H]thymidine incorporation. Results shown are the mean ± SD of the group (n = 3) and are representative of two independent experiments. #, p < 0.05 for significant difference between CD4+CD25− and CD4+CD25high cultures. ∗, p < 0.05 for significant difference between CD4+CD25− and CD4+CD25− and CD4+CD25high cocultures.

Reduced numbers of effector T cells and suppressed IFN-γ production in anti-CD25 Ab-treated T. gondii-infected mice

Numerous studies have shown that anti-CD25 Ab (clone PC61) treatment leaves mice essentially devoid of cells expressing CD25 for an extended period (27, 32), an observation we confirm in this study during T. gondii infection (Fig. 4⇓); CD25 expressing splenic CD4+ T cells were almost completely undetectable following anti-CD25 Ab administration (detection day 9 postinfection with Ab injected i.p. 4 days before infection) (Fig. 4⇓A). These results were largely reproduced when examining mesenteric lymph node cells (data not shown). Importantly, and consistent with previous reports (27, 32), PC61 administration failed to induce complete depletion of Foxp3+ Tregs, instead promoting a maximal 40% reduction in Foxp3-expressing cell numbers (Fig. 4⇓, B and C). As we have demonstrated that during acute stage T. gondii infection the overwhelming majority of CD4CD25high cells display effector rather than regulatory characteristics, these results directly indicate that anti-CD25 Abs primarily deplete effector T cells during T. gondii infection. In agreement, anti-CD25 Abs principally targeted activated T cell populations, as shown by the significant decrease in the number of CD4+ T cells expressing high levels of the activation marker CD44 following PC61 administration (28.9 ± 11.6 × 106 in control mAb-treated vs 13.3 ± 4.5 × 106 in anti-CD25-treated) (Fig. 4⇓A).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Anti-CD25 administration depletes activated CD25+ T cells and significantly reduces IFN-γ production. Groups of five B6 males were administered 1 mg of anti-CD25 (clone PC61) or control mAb (clone HRPN) i.p. 5 days before oral infection with 10 ME49 cysts. A, The number of splenic-activated CD4+ T cells were calculated on day 9 postinfection by measuring CD25 and CD44 expression. CD25 detection was performed using the 7D4 clone anti-CD25 Ab. B, Representative dot plots showing Foxp3 expression on splenic CD4+ T cells on day 8 postinfection. C, The number of splenic CD4+Foxp3+ cells on day 8 postinfection in control Ab- and anti-CD25 Ab-treated mice. D–F, The level of IFN-γ and IL-10 in PC61- and HRPN-treated mice was determined on day 9 postinfection in plasma (D) by ELISA and in liver (E and F) by real-time PCR (Taqman). Results shown are mean ± SD of the group (n = 5) and are representative of two independent experiments. ∗, p < 0.05; ∗∗, p < 0.01.

Consequently, to examine whether the depressed effector T cell responses observed during T. gondii infection following anti-CD25 Ab administration correlated with a functional defect in proinflammatory immune responses, we next determined the levels of the prototypic type I cytokine IFN-γ in T. gondii-infected mice treated with anti-CD25 Abs. Our results show that PC61 administration reduced systemic plasma IFN-γ levels (detected on day 8 postinfection) compared with control Ab-treated infected mice (Fig. 4⇑D). Although the reduction in IFN-γ production failed to reach statistical significance in any individual experiments, a reproducible reduction in IFN-γ production was observed in four separate experiments, with the percentage of reductions relative to controls of 11, 20, 27, and 52%. Furthermore, the reduced serum IFN-γ levels were paralleled by significantly reduced levels of IFN-γ mRNA in the liver of anti-CD25 Ab-treated mice on day 8 postinfection (Fig. 4⇑E).

CD4+ T cells, specifically Th1 cells that express T-bet and coproduce IFN-γ, have also been shown to be the primary source of host-protective IL-10 during T. gondii infection (33). Importantly, however, the expression level of IL-10 mRNA in the liver was comparable in anti-CD25 Ab- and control-treated mice (Fig. 4⇑F), indicating that the regulatory IL-10 response was largely intact in anti-CD25 Ab-treated mice. Combined, these data definitively demonstrate that in vivo administration of anti-CD25 Abs can negatively effect proinflammatory immune responses during an inflammatory disorder, and that CD25 expression is up-regulated on T. gondii-specific activated effector T cells, which are required for effective proinflammatory type 1 responses.

Anti-CD25 Ab administration suppresses IFN-γ production by CD4+ T cells

To examine the T cell populations affected by anti-CD25 Ab administration leading to impaired IFN-γ expression, we used IFN-γ bicistronic reporter mice (YETI) (25), enabling the detection and quantification of cell-specific IFN-γ production directly ex vivo, negating the requirement for subsequent restimulation in vitro. Importantly, yellow fluorescent protein (yFP) expression in YETI mice not only marks cells actively producing IFN-γ, but also marks cells that have expressed IFN-γ, providing an accurate representation of both current and historical cellular function (25). As expected, the majority of CD4+ T cells in naive mice were found to be yFP-negative (25), indicating that few CD4+ T cells were polarized type 1 effector/memory cells (Fig. 5⇓A). The CD4+ yFP-positive cells found in liver, spleen, blood, and mesenteric lymph nodes in naive mice displayed heterogeneity in CD25 expression, suggesting the existence of a number of different populations of IFN-γ-producing CD4+ T cells at various stages of cellular activation. All yFP-positive cells irrespective of CD25 expression or tissue/organ location were, however, uniformly CD44high (results not shown).

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

Anti CD25 Ab administration suppresses IFN-γ production by CD4+ T cells. Groups of three bicistronic IFN-γ-enhanced yFP (IFN-γ-yFP) reporter male mice (Yeti) were given 1 mg of anti-CD25 (clone PC61) or control mAb (clone HRPN) i.p. 5 days before oral infection with brain homogenate containing 10 ME49 cysts or sham uninfected brain homogenate. A, Representative dot plots showing yFP expression relative to CD25 expression (using 7D4 to detect) on CD4+ T cells from spleen, mesenteric lymph, liver, and blood on day 8 postinfection. B, Relative frequency of CD4+ T cells expressing yFP in anti-CD25 Ab-treated and control Ab-treated T. gondii-infected and sham-infected mice on day 8 postinfection. Results shown are mean ± SD of the group (n = 3) and are representative of two independent experiments. ∗, p < 0.05.

In agreement with Fig. 2⇑, up-regulation of CD25 on CD4+ T cells was observed on day 8 postinfection in all tissues examined and this response correlated with significant up-regulation of yFP expression; importantly in all the tissues examined the majority of yFP-expressing CD4+ T cells coexpressed CD25 (Fig. 5⇑A). Thus, virtually all CD4+CD25high cells derived from liver and blood were yFP-positive, marking CD4CD25high cells as predominantly effector rather than Tregs in these tissues during acute T. gondii infection. In contrast, a number of CD4+CD25high cells derived from spleen and mesenteric lymph nodes of T. gondii-infected mice did not coexpress yFP, suggesting that these cells are either Tregs or activated T cells that have failed to produce IFN-γ (Fig. 5⇑A). Irrespective of this finding, our results in Fig. 3⇑ clearly show that splenic CD4+CD25high T cells predominantly exert effector rather than regulatory functionality during acute stage T. gondii infection.

In support of our hypothesis that anti-CD25 Ab administration directly suppresses proinflammatory protective immune responses during acute T. gondii infection, we observed significantly reduced frequency of yFP-positive CD4+ T cells in the liver, mesenteric lymph node, blood, and spleen of T. gondii-infected mice treated with anti-CD25 Abs as compared with control Ab-treated infected mice (Fig. 5⇑B). Similarly, significantly reduced frequency of yFP-positive CD8+ T cells were observed in the liver of T. gondii-infected mice treated with anti-CD25 Abs as compared with control Ab-treated infected mice (results not shown). In contrast anti-CD25 Ab administration did not significantly reduce yFP expression by CD8+ T cells in the spleen, mesenteric lymph node, or blood, suggesting that effector CD8+ T cells are less affected by anti-CD25 Ab than CD4+ T cells (results not shown).

Heterogeneous expression of yFP by CD4+ T cells occurs during T. gondii infection, as shown in infected, isotype control group (Fig. 5⇑) and as published in Mayer et al. (34). Importantly, the expression level of yFP is directly correlated to the capacity to produce IFN-γ following in vitro restimulation, with cells expressing highest levels of yFP capable of producing most IFN-γ protein (34). Interestingly, the highest level of yFP expression was routinely observed in CD4+ T cells expressing highest levels of CD25 (Fig. 5⇑A), marking these potent IFN-γ-producing cells as principal targets of anti-CD25 Abs. Thus, anti-CD25 Ab administration significantly reduced the yFPhigh CD4+ T cell subsets in the liver and blood. A reduction in the frequency of yFPhigh CD4+ T cells was also observed in the mesenteric lymph node and spleen, but the reduction failed to reach statistical significance (results not shown). Combined, these results demonstrate that anti-CD25 Abs can directly target effector IFN-γ-producing CD4+ T cells during acute T. gondii infection leading to systemic reductions in IFN-γ production and impaired protective T cell immunity.

Anti-CD25 Ab administration does not impair resistance to chronic stage T. gondii infection

It is well established that anti-parasitic CD4+ T cells and CD8+ T cells are required for the control of parasite reactivation in the brain and prevention of Toxoplasma encephalitis during chronic T. gondii infection (22). However, the activation status of these CD4+ and CD8+ T cells is relatively unknown. Consequently it is unclear whether protective CD4+ and CD8+ T cells in the spleen or brain express CD25 during chronic infection, and whether these cells may also be affected by anti-CD25 Ab administration. Additionally, it has previously been shown that natural Tregs that coexpress Foxp3 and CD25 can suppress protective immunity during a number of infections, including Leishmania major infection, enabling parasite persistence and the establishment of chronic infections (35, 36). Thus, CD25-expressing CD4+ T cells may also play an important regulatory role during chronic T. gondii infection, lowering proinflammatory responses, moderating immune-mediated pathology but impairing parasite control. As a result, anti-CD25 Ab administration during chronic T. gondii infection could potentially augment anti-parasitic immune responses and enhance parasite control. In support of this hypothesis, Foxp3 mRNA levels in the brain were increased during chronic infection compared with uninfected control levels (results not shown). Moreover, we have shown that CD25 up-regulation on effector T cells is transient during T. gondii infection and occurs primarily during the acute phase of infection, suggesting that CD4+CD25high cells are functionally disparate during acute and chronic stages of T. gondii infection (Fig. 2⇑). In agreement with a regulatory rather than effector role of CD25-expressing cells during chronic infection, CD4+CD25high cells isolated from both brain and spleen during chronic infection failed to produce significant quantities of IFN-γ following T. gondii antigenic restimulation (as compared with CD4+CD25− cells) and splenic CD4+CD25high cells moderately suppressed IFN-γ production by CD4+CD25− cells (Fig. 6⇓, A and B).

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

Anti-CD25 Ab administration does not affect control of chronic T. gondii infection. A and B, CD4+CD25− and CD4+CD25high T cells were sort purified from spleen (A) or brain (B) of mice chronically infected with T. gondii (day 50 postinfection). CD4+CD25− and CD4+CD25high cells were cultured with T. gondii-pulsed APC for 60 h and IFN-γ production was assayed in supernatant by ELISA. Results shown are the mean ± SD of the group (n = 3) and are representative of two independent experiments. #, p < 0.05 for significant difference between CD4+CD25− and CD4+CD25high cultures. ∗, p < 0.05 for significant difference between CD4+CD25− and CD4+CD25− and CD4+CD25high cocultures. C–F, The 1 mg of anti-CD25 Ab (clone PC61) or control Ab was administered to mice chronically infected with T. gondii (day 56 postinfection). C, At 5 days postantibody administration, the number of total brain CD4+ T cells, CD4CD25high (using 7D4 to detect), and CD4+Foxp3+ cells were enumerated to determine the effectiveness of anti-CD25 Ab-mediated depletion. ∗, p < 0.05 for significant difference between control Ab- and anti-CD25 Ab-treated groups. Weight loss (D) and survival (E) were followed following Ab administration. Weight values are expressed as the percentage weight relative to preantibody administration (day 56 weights). ∗, p < 0.05 for significant difference between control Ab- and anti-CD25 Ab-treated groups. F, The effect of anti-CD25 Ab administration on brain parasite control and immune responses were determined 30 days postantibody administration by quantifying parasite (tachyzoite) and IFN-γ levels by real-time PCR. Results are shown as fold change relative to control Ab-treated mice. None of the differences were statistically significant in this or a replicate experiment.

To directly determine the relative importance of CD25-expressing natural Tregs during chronic T. gondii infection, we administered anti-CD25 Abs to chronically infected mice (day 56 postinfection) and determined whether Ab administration significantly modulated disease control. Anti-CD25 Ab administration effectively neutralized CD25 expression in the spleen (results not shown) and brain (Fig. 6⇑C) 5 and 10 days postadministration (days 5 and 10 postadministration were examined in separate experiments with results from day 5 postadministration shown). However, Foxp3 levels were not significantly reduced in anti-CD25 Ab-treated mice (Fig. 6⇑C). Importantly, it has previously been shown that Foxp3+ cells that survive anti-CD25 Ab treatment are functionally inactive (32), indicating that anti-CD25 Ab administration should still ablate Treg functionality during chronic T. gondii infection even if all Foxp3+ T cells are not eliminated. Nonetheless, Ab administration did not significantly alter the level of weight loss (relative to preantibody administration baseline levels) or mortality over a 30-day period posttreatment (Fig. 6⇑, D and E) and this correlated with unaltered parasite control (measuring invasive tachyzoite forms of parasite) and IFN-γ production in the brain of anti-CD25 Ab-treated mice compared with control treated groups (Fig. 6⇑F). Combined these results demonstrate that CD25-expressing natural Tregs do not suppress anti-parasitic immune responses during chronic T. gondii infection and are not required to limit excessive inflammation. In support of this conclusion, the frequency of CD4+Foxp3+ cells in the brain (of total CD4+ T cells) was not elevated in chronically infected mice compared with naive mice. Thus, the increase in brain Foxp3 mRNA expression during chronic infection is due to the increase in total CD4+ T cells that migrate to and reside in brains of chronically infected mice, rather than specific localization and migration of Tregs to the site of infection, as is observed during L. major infection (35).

Discussion

In this study, we demonstrate that during acute T. gondii infection transient up-regulation of CD25 expression occurs on CD4+ T cells during the expansion phase of the T cell response. Importantly, the vast majority of CD4+CD25high cells generated during acute T. gondii infection do not coexpress Foxp3, clearly defining the cells as effector cells rather than Tregs. Anti-CD25 Abs directly target these CD25-expressing effector CD4+ T cell populations leading to impaired proinflammatory immune responses, exemplified by significantly reduced T cell-derived IFN-γ production. Thus anti-CD25 Ab administration initially reduced the severity of T cell-dependent immune-mediated pathology during early acute T. gondii infection, but led to the failure of parasite control and ultimately death in a large number of anti-CD25 Ab-treated mice. In contrast, anti-CD25 Ab administration did not affect parasite control during chronic T. gondii infection, suggesting that CD25-expressing T cells do not significantly regulate anti-parasitic immunity during the later chronic stage of T. gondii infection.

The results observed in this study are entirely consistent with a direct effect of anti-CD25 Ab administration on proinflammatory effector T cell populations. Thus, although (as expected) anti-CD25 Ab administration reduced the number of Foxp3+ natural Tregs, it also reduced circulating IFN-γ levels (in all four experiments examined) and resulted in significantly reduced IFN-γ mRNA levels in the liver. Using IFN-γ yFP reporter (YETI) mice, we subsequently showed that anti-CD25 Ab administration leads to reduced IFN-γ production by CD4+ T cells in the liver, spleen, blood, and mesenteric lymph node. Consequently our results differ from those obtained by Lund et al. (37), where specific natural Treg depletion reduced localized proinflammatory immune responses at the mucosal site of HSV infection, but enhanced proinflammatory immune responses in tissue draining lymph nodes. Therefore, whereas in their model, the specific depletion of natural Tregs (performed in Foxp3 diphtheria toxoid receptor transgenic mice) inhibited effector T cell migration to nonlympoid tissue sites of infection, we show that anti-CD25 Ab administration promoted systemic suppression of effector T cell responses. Combined, these data demonstrate the lack of specificity of anti-CD25 Abs to target natural Tregs compared with the more recently generated Foxp3-dtr system (38).

It has previously been demonstrated that effector CD4+ T cells are responsible for the onset of immune-mediated pathology and severe weight loss in B6 mice following oral T. gondii infection. (20, 31, 39, 40). Consistent with this demonstration, anti-CD25 Ab administration modulated body weight loss during acute T. gondii infection and significantly reduced the severity of liver pathology on day 8 of infection, indicating a direct effect on effector T cell populations. Nevertheless, it has also previously been reported that Th1 CD4+ T cells that coproduce IFN-γ are the primary source of host-protective IL-10 during T. gondii infection (33). Consequently, given the effect of anti-CD25 Ab administration on CD4+ T cell-derived IFN-γ production, it was foreseeable that Ab administration could also have impaired IL-10 production, and that this secondary Ab effect may have contributed to the enhanced susceptibility of anti-CD25 Ab-treated mice during acute T. gondii infection. Our results show that IL-10 mRNA levels were not significantly reduced in the liver following anti-CD25 Ab administration, suggesting that CD25 expression is not critical for the production of host-protective IL-10 during acute T. gondii infection. These results appear to further underline the effects of anti-CD25 Ab administration on effector proinflammatory responses, rather than regulatory based responses during acute T. gondii infection. As we did not specifically examine the cellular source or protein levels of IL-10 additional experiments are, however, required to determine the extent that anti-CD25 Ab treatment alters IL-10 production by Th1 CD4+ T cells during infection. Interestingly, although we highlight the importance of CD25 expression on effector T cells during the acute stage of primary T. gondii infection, administration of anti-CD25 Abs did not affect vaccine-induced protection during secondary challenge infection. Thus, anti-CD25 Ab administration to mice previously infected with the noncyst forming attenuated TS-4 strain of parasites did not inhibit protective immune responses and anti-CD25 Ab- and control-treated vaccinated mice displayed equivalent ability to prevent brain cyst development (results not shown). These results indicate that CD25-expressing effector T cell populations are not critically required for vaccine-induced protection to T. gondii infection.

Despite the pathological role of CD4+ T cells during early acute T. gondii infection, it is well established that effector CD4+ T cells and CD8+ T cells are required for protection and parasite control during late acute and chronic stages of infection (41, 42, 43). Consequently, as anti-CD25 Ab treatment reduced the number of activated (CD44high) CD4+ spleen cells by more than 50% on day 9 of infection, it is not surprising that anti-CD25 Ab treated mice displayed elevated parasite levels in the ileum on days 8 and 15 postinfection, exhibited more brain cysts on day 30 post infection and succumbed more rapidly to late/acute and chronic stage infection. Nonetheless, we have also demonstrated that anti-CD25 Ab administration during established chronic stage of T. gondii infection did not affect brain-localized parasite control or anti-T. gondii immunity, suggesting that anti-CD25 Abs do not negatively affect protective T cell responses at this stage. In agreement, transient expression of CD25 on effector T cells was observed only during acute T. gondii infection and CD25 expression on splenic CD4+ and CD8+ T cells during chronic stages of infection was comparable to that observed in naive mice. Moreover, brain infiltrating CD4+ T cell populations did not express higher levels of CD25 than splenic CD4+ T cells during chronic T. gondii infection, indicating that CD25 expressing lymphocytes do not preferentially migrate to and reside at the local site of parasite persistence during chronic infection (data not shown). In addition, CD4+CD25high cells isolated from the spleen and brain of chronically infected mice displayed regulatory rather than effector phenotypes following in vitro Ag-specific stimulation. Thus, CD25-expressing CD4+ T cells are primarily regulatory rather than effector cells during chronic T. gondii infection and effector CD4+ and CD8+ T cells do not require CD25 expression to mediate parasite control during the later stages of infection. Importantly, these results also demonstrate that natural Tregs do not control anti-parasitic immune responses during chronic T. gondii infection, as administration of anti-CD25 Abs did not augment parasite control. Consequently, it is unlikely that natural Tregs are the source of IL-10 that is required for the limitation of immune-mediated pathology during chronic T. gondii infection (44).

Most studies that use anti-CD25 Ab administration to investigate Tregs assume that the Ab affects only that subset and, for the most part, these studies have disregarded effects on cells other than Tregs that can up-regulate CD25 in response to infection. This decision is surprising because Ag-stimulated effector T cells (and others) up-regulate CD25 and IL-2 drives T cell proliferation in response to Ag stimulation. Notably, anti-CD25 Abs have been used clinically to dampen autoimmunity and reduce rejection of transplants (45), uses that run counter to the notion that anti-CD25 augments immunity. The reason for the pronounced effect of anti-CD25 Ab on effector T cell populations during acute T. gondii infection, but not in other models is unclear, but is likely correlated with the extreme polarized systemic proinflammatory immune response that develops during T. gondii infection. As CD25 is up-regulated on a significant percentage of the CD4+ and CD8+ T cell population during acute T. gondii infection, it is foreseeable that any effect of anti-CD25 Ab treatment on effector T cell populations is more pronounced during acute T. gondii infection, and might not be as apparent in other models.

In conclusion, we have shown that during acute T. gondii infection, anti-CD25 Abs directly target effector CD4+ T cell populations, significantly modulating the proinflammatory immune response to T. gondii infection. Although initially this limits immune-mediated pathology, it also leads to the failure to control parasite dissemination and encystment and finally death. Thus, although highlighting the role of CD25 expressing T cells for the control of acute T. gondii infection, we importantly also demonstrate the significant limitations of using anti-CD25 Abs in vivo during inflammatory diseases to investigate the importance of Tregs. On the basis of our findings, we propose that studies that use anti-CD25 Abs during inflammatory settings should take note of the effects on the entire T cell population, not solely those expressing markers associated with Tregs.

Acknowledgments

We thank Katja Mohrs for technical assistance. We also thank Dr. Julius Hafalla and Dr. Brian de Souza for critical reading of the manuscript.

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported by a Public Health Service Grant AI061587 from the National Institute of Allergy and Infectious Diseases and by funds provided by the Trudeau Institute.

  • ↵2 Address correspondence and reprint requests to Dr. Kevin N. Couper at the current address: Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K. E-mail address: kevin.couper{at}lshtm.ac.uk

  • ↵3 Abbreviations used in this paper: Treg, regulatory T cell; yFP, yellow fluorescent protein.

  • Received September 16, 2008.
  • Accepted January 18, 2009.
  • Copyright © 2009 by The American Association of Immunologists, Inc.

References

  1. ↵
    Belkaid, Y.. 2007. Regulatory T cells and infection: a dangerous necessity. Nat. Rev. Immunol. 7: 875-888.
    OpenUrlCrossRefPubMed
  2. ↵
    Sakaguchi, S., M. Ono, R. Setoguchi, H. Yagi, S. Hori, Z. Fehervari, J. Shimizu, T. Takahashi, T. Nomura. 2006. Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol. Rev. 212: 8-27.
    OpenUrlCrossRefPubMed
  3. ↵
    Waldmann, T. A.. 1986. The multichain interleukin-2 receptor: from the gene to the bedside. Harvey Lect. 82: 1-17.
    OpenUrlPubMed
  4. ↵
    Fontenot, J. D., J. P. Rasmussen, L. M. Williams, J. L. Dooley, A. G. Farr, A. Y. Rudensky. 2005. Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity 22: 329-341.
    OpenUrlCrossRefPubMed
  5. ↵
    Suvas, S., U. Kumaraguru, C. D. Pack, S. Lee, B. T. Rouse. 2003. CD4+CD25+ T cells regulate virus-specific primary and memory CD8+ T cell responses. J. Exp. Med. 198: 889-901.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Aseffa, A., A. Gumy, P. Launois, H. R. MacDonald, J. A. Louis, F. Tacchini-Cottier. 2002. The early IL-4 response to Leishmania major and the resulting Th2 cell maturation steering progressive disease in BALB/c mice are subject to the control of regulatory CD4+CD25+ T cells. J. Immunol. 169: 3232-3241.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Mendez, S., S. K. Reckling, C. A. Piccirillo, D. Sacks, Y. Belkaid. 2004. Role for CD4+CD25+ regulatory T cells in reactivation of persistent leishmaniasis and control of concomitant immunity. J. Exp. Med. 200: 201-210.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Hisaeda, H., Y. Maekawa, D. Iwakawa, H. Okada, K. Himeno, K. Kishihara, S. Tsukumo, K. Yasutomo. 2004. Escape of malaria parasites from host immunity requires CD4+CD25+ regulatory T cells. Nat. Med. 10: 29-30.
    OpenUrlCrossRefPubMed
  9. ↵
    Taylor, M. D., L. LeGoff, A. Harris, E. Malone, J. E. Allen, R. M. Maizels. 2005. Removal of regulatory T cell activity reverses hyporesponsiveness and leads to filarial parasite clearance in vivo. J. Immunol. 174: 4924-4933.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Nardelli, D. T., T. F. Warner, S. M. Callister, R. F. Schell. 2006. Anti-CD25 antibody treatment of mice vaccinated and challenged with Borrelia spp. does not exacerbate arthritis but inhibits borreliacidal antibody production. Clin. Vaccine Immunol. 13: 884-891.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Dittmer, U., H. He, R. J. Messer, S. Schimmer, A. R. Olbrich, C. Ohlen, P. D. Greenberg, I. M. Stromnes, M. Iwashiro, S. Sakaguchi, et al 2004. Functional impairment of CD8+ T cells by regulatory T cells during persistent retroviral. Infect. Immun. 20: 293-303.
    OpenUrl
  12. ↵
    Kotner, J., R. Tarleton. 2007. Endogenous CD4+CD25+ regulatory T cells have a limited role in the control of Trypanosoma cruzi infection in mice. Infect. Immun. 75: 861-869.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Couper, K. N., D. G. Blount, M. S. Wilson, J. C. Hafalla, Y. Belkaid, M. Kamanaka, R. A. Flavell, J. B. de Souza, E. M. Riley. 2008. IL-10 from CD4CD25Foxp3CD127 adaptive regulatory T cells modulates parasite clearance and pathology during malaria infection. PLoS Pathog. 4: e1000004
    OpenUrlCrossRefPubMed
  14. ↵
    Gazzinelli, R. T., S. Hieny, T. A. Wynn, S. Wolf, A. Sher. 1993. Interleukin 12 is required for the T-lymphocyte-independent induction of interferon γ by an intracellular parasite and induces resistance in T-cell-deficient hosts. Proc. Natl. Acad. Sci. USA 90: 6115-6119.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Bennouna, S., S. K. Bliss, T. J. Curiel, E. Y. Denkers. 2003. Cross-talk in the innate immune system: neutrophils instruct recruitment and activation of dendritic cells during microbial infection. J. Immunol. 171: 6052-6058.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Bliss, S. K., L. C. Gavrilescu, A. Alcaraz, E. Y. Denkers. 2001. Neutrophil depletion during Toxoplasma gondii infection leads to impaired immunity and lethal systemic pathology. Infect. Immun. 69: 4898-4905.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Hunter, C. A., C. S. Subauste, V. H. Van Cleave, J. S. Remington. 1994. Production of γ interferon by natural killer cells from Toxoplasma gondii-infected SCID mice: regulation by interleukin-10, interleukin-12, and tumor necrosis factor alpha. Infect. Immun. 62: 2818-2824.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Johnson, L. L., F. P. VanderVegt, E. A. Havell. 1993. Gamma interferon-dependent temporary resistance to acute Toxoplasma gondii infection independent of CD4+ or CD8+ lymphocytes. Infect. Immun. 61: 5174-5180.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Mordue, D. G., L. D. Sibley. 2003. A novel population of Gr-1+-activated macrophages induced during acute toxoplasmosis. J. Leukocyte Biol. 74: 1015-1025.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Liesenfeld, O., J. Kosek, J. S. Remington, Y. Suzuki. 1996. Association of CD4+ T cell-dependent, interferon-γ-mediated necrosis of the small intestine with genetic susceptibility of mice to peroral infection with Toxoplasma gondii. J. Exp. Med. 184: 597-607.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Denkers, E. Y., R. T. Gazzinelli. 1998. Regulation and function of T-cell-mediated immunity during Toxoplasma gondii infection. Clin. Microbiol. Rev. 11: 569-588.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Gazzinelli, R., Y. Xu, S. Hieny, A. Cheever, A. Sher. 1992. Simultaneous depletion of CD4+ and CD8+ T lymphocytes is required to reactivate chronic infection with Toxoplasma gondii. J. Immunol. 149: 175-180.
    OpenUrlAbstract
  23. ↵
    Johnson, L. L.. 1992. SCID mouse models of acute and relapsing chronic Toxoplasma gondii infections. Infect. Immun. 60: 3719-3724.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Johnson, L. L.. 1992. A protective role for endogenous tumor necrosis factor in Toxoplasma gondii infection. Infect. Immun. 60: 1979-1983.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Stetson, D. B., M. Mohrs, R. L. Reinhardt, J. L. Baron, Z. E. Wang, L. Gapin, M. Kronenberg, R. M. Locksley. 2003. Constitutive cytokine mRNAs mark natural killer (NK) and NK T cells poised for rapid effector function. J. Exp. Med. 198: 1069-1076.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Johnson, L. L., K. N. Berggren, F. M. Szaba, W. Chen, S. T. Smiley. 2003. Fibrin-mediated protection against infection-stimulated immunopathology. J. Exp. Med. 197: 801-806.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Couper, K. N., D. G. Blount, J. B. de Souza, I. Suffia, Y. Belkaid, E. M. Riley. 2007. Incomplete depletion and rapid regeneration of Foxp3+ regulatory T cells following anti-CD25 treatment in malaria-infected mice. J. Immunol. 178: 4136-4146.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Johnson, L. L., P. Lanthier, J. Hoffman, W. Chen. 2004. Vaccination protects B cell-deficient mice against an oral challenge with mildly virulent Toxoplasma gondii. Vaccine 22: 4054-4061.
    OpenUrlCrossRefPubMed
  29. ↵
    Kasper, L. H., M. S. Bradley, E. R. Pfefferkorn. 1984. Identification of stage-specific sporozoite antigens of Toxoplasma gondii by monoclonal antibodies. J. Immunol. 132: 443-449.
    OpenUrlAbstract
  30. ↵
    Lindgren, H., S. Stenmark, W. Chen, A. Tarnvik, A. Sjostedt. 2004. Distinct roles of reactive nitrogen and oxygen species to control infection with the facultative intracellular bacterium Francisella tularensis. Infect. Immun. 72: 7172-7182.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Smiley, S. T., P. A. Lanthier, K. N. Couper, F. M. Szaba, J. E. Boyson, W. Chen, L. L. Johnson. 2005. Exacerbated susceptibility to infection-stimulated immunopathology in CD1d-deficient mice. J. Immunol. 174: 7904-7911.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Kohm, A. P., J. S. McMahon, J. R. Podojil, W. S. Begolka, M. DeGutes, D. J. Kasprowicz, S. F. Ziegler, S. D. Miller. 2006. Cutting edge: anti-CD25 monoclonal antibody injection results in the functional inactivation, not depletion, of CD4+CD25+ T regulatory cells. J. Immunol. 176: 3301-3305.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Jankovic, D., M. C. Kullberg, C. G. Feng, R. S. Goldszmid, C. M. Collazo, M. Wilson, T. A. Wynn, M. Kamanaka, R. A. Flavell, A. Sher. 2007. Conventional T-bet+Foxp3− Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection. J. Exp. Med. 204: 273-283.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Mayer, K. D., K. Mohrs, S. R. Crowe, L. L. Johnson, P. Rhyne, D. L. Woodland, M. Mohrs. 2005. The functional heterogeneity of type 1 effector T cells in response to infection is related to the potential for IFN-γ production. J. Immunol. 174: 7732-7739.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Belkaid, Y., C. A. Piccirillo, S. Mendez, E. M. Shevach, D. L. Sacks. 2002. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature 420: 502-507.
    OpenUrlCrossRefPubMed
  36. ↵
    Belkaid, Y., R. B. Blank, I. Suffia. 2006. Natural regulatory T cells and parasites: a common quest for host homeostasis. Immunol. Rev. 212: 287-300.
    OpenUrlCrossRefPubMed
  37. ↵
    Lund, J. M., L. Hsing, T. T. Pham, A. Y. Rudensky. 2008. Coordination of early protective immunity to viral infection by regulatory T cells. Science 320: 1220-1224.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Kim, J. M., J. P. Rasmussen, A. Y. Rudensky. 2007. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8: 191-197.
    OpenUrlCrossRefPubMed
  39. ↵
    Liesenfeld, O., H. Kang, D. Park, T. A. Nguyen, C. V. Parkhe, H. Watanabe, T. Abo, A. Sher, J. S. Remington, Y. Suzuki. 1999. TNF-α, nitric oxide and IFN-γ are all critical for development of necrosis in the small intestine and early mortality in genetically susceptible mice infected perorally with Toxoplasma gondii. Parasite Immunol. 21: 365-376.
    OpenUrlCrossRefPubMed
  40. ↵
    Suzuki, Y., A. Sher, G. Yap, D. Park, L. E. Neyer, O. Liesenfeld, M. Fort, H. Kang, E. Gufwoli. 2000. IL-10 is required for prevention of necrosis in the small intestine and mortality in both genetically resistant BALB/c and susceptible C57BL/6 mice following peroral infection with Toxoplasma gondii. J. Immunol. 164: 5375-5382.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Araujo, F. G.. 1991. Depletion of L3T4+ (CD4+) T lymphocytes prevents development of resistance to Toxoplasma gondii in mice. Infect. Immun. 59: 1614-1619.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Araujo, F. G.. 1992. Depletion of CD4+ T cells but not inhibition of the protective activity of IFN-γ prevents cure of toxoplasmosis mediated by drug therapy in mice. J. Immunol. 149: 3003-3007.
    OpenUrlAbstract
  43. ↵
    Johnson, L. L., P. C. Sayles. 2002. Deficient humoral responses underlie susceptibility to Toxoplasma gondii in CD4-deficient mice. Infect. Immun. 70: 185-191.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Wilson, E. H., U. Wille-Reece, F. Dzierszinski, C. A. Hunter. 2005. A critical role for IL-10 in limiting inflammation during toxoplasmic encephalitis. J. Neuroimmunol. 165: 63-74.
    OpenUrlCrossRefPubMed
  45. ↵
    Morris, J. C., T. A. Waldmann. 2000. Advances in interleukin 2 receptor targeted treatment. Ann. Rheum. Dis. 59: (Suppl. 1):109-114.
    OpenUrlCrossRef
PreviousNext
Back to top

In this issue

The Journal of Immunology: 182 (7)
The Journal of Immunology
Vol. 182, Issue 7
1 Apr 2009
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Anti-CD25 Antibody-Mediated Depletion of Effector T Cell Populations Enhances Susceptibility of Mice to Acute but Not Chronic Toxoplasma gondii Infection
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Anti-CD25 Antibody-Mediated Depletion of Effector T Cell Populations Enhances Susceptibility of Mice to Acute but Not Chronic Toxoplasma gondii Infection
Kevin N. Couper, Paula A. Lanthier, Georgia Perona-Wright, Lawrence W. Kummer, Wangxue Chen, Stephen T. Smiley, Markus Mohrs, Lawrence L. Johnson
The Journal of Immunology April 1, 2009, 182 (7) 3985-3994; DOI: 10.4049/jimmunol.0803053

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Anti-CD25 Antibody-Mediated Depletion of Effector T Cell Populations Enhances Susceptibility of Mice to Acute but Not Chronic Toxoplasma gondii Infection
Kevin N. Couper, Paula A. Lanthier, Georgia Perona-Wright, Lawrence W. Kummer, Wangxue Chen, Stephen T. Smiley, Markus Mohrs, Lawrence L. Johnson
The Journal of Immunology April 1, 2009, 182 (7) 3985-3994; DOI: 10.4049/jimmunol.0803053
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • FAR 889
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2022 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606