Skip to main content
Infection and Immunity logoLink to Infection and Immunity
. 2008 Mar 3;76(5):1908–1919. doi: 10.1128/IAI.01233-07

Functional Analysis of Effector and Regulatory T Cells in a Parasitic Nematode Infection

Sebastian Rausch 1, Jochen Huehn 2, Dennis Kirchhoff 3, Justyna Rzepecka 1, Corinna Schnoeller 1, Smitha Pillai 1, Christoph Loddenkemper 4, Alexander Scheffold 3, Alf Hamann 2, Richard Lucius 1, Susanne Hartmann 1,*
PMCID: PMC2346705  PMID: 18316386

Abstract

Parasitic nematodes typically modulate T-cell reactivity, primarily during the chronic phase of infection. We analyzed the role of CD4-positive (CD4+) T effector (Teff) cells and regulatory T (Treg) cells derived from mice chronically infected with the intestinal nematode Heligmosomoides polygyrus. Different CD4+ T-cell subsets were transferred into naïve recipients that were subsequently infected with H. polygyrus. Adoptive transfer of conventional Teff cells conferred protection and led to a significant decrease in the worm burdens of H. polygyrus-infected recipients. Roughly 0.2% of the CD4+ T cells were H. polygyrus specific based on expression of CD154, and cells producing interleukin 4 (IL-4) and IL-13 were highly enriched within the CD154+ population. In contrast, adoptive transfer of Treg cells, characterized by the markers CD25 and CD103 and the transcription factor Foxp3, had no effect on the worm burdens of recipients. Further analysis showed that soon after infection, the number of Foxp3+ Treg cells temporarily increased in the inflamed tissue while effector/memory-like CD103+ Foxp+ Treg cells systemically increased in the draining lymph nodes and spleen. In addition, Treg cells represented a potential source of IL-10 and reduced the expression of IL-4. Finally, under in vitro conditions, Treg cells from infected mice were more potent suppressors than cells derived from naïve mice. In conclusion, our data indicate that small numbers of Teff cells have the ability to promote host protective immune responses, even in the presence of Treg cells.


Infections with parasitic nematodes have been shown to markedly modulate the host's immune system as a means to escape immune effector mechanisms and to ensure parasite survival within an immunocompetent host (35, 55). Highly effective immune evasion mechanisms are key elements of the long-lasting persistence of parasitic worms. Both parasite-specific and nonspecific immune suppression are well documented (19, 47, 60). Usually, acute infection stimulates antigen-specific T-cell proliferation, but with increasing exposure to parasite antigens, the immune system becomes hyporesponsive (34).

Immune responses against gastrointestinal nematodes include strong immunoglobulin G1 (IgG1) and IgE responses, eosinophilia, intestinal mastocytosis, goblet cell hyperplasia, and smooth-muscle hypercontractility (1, 12, 70). Th2-driven effector mechanisms were shown to be protective against gastrointestinal helminths, whereby interleukin 13 (IL-13) and IL-4 play an important role in primary, as well as secondary, infections (7, 13, 15, 53). Recently, alternatively activated macrophages have been introduced as an IL-4-dependent effector population essential for protective immunity to challenge infections with Heligmosomoides polygyrus (4). However, there is a lack of information on the activity of CD4-positive (CD4+) T-cell subsets during the chronic phase of primary nematode infections (when T-cell reactivity is suppressed) and on the T-cell subsets that contribute to host-protective or parasite-beneficial immune responses. In contrast, the interplay of T-cell subsets and regulation of effector responses in bacterial (28) and protozoan infections, like those with Leishmania (9, 38, 66) and Plasmodium (21, 44) species, are well defined.

It has been suggested that nematode infections reduce both Th1- and Th2-mediated responses by profoundly influencing regulatory pathways (5, 35). Regulatory T (Treg) cells represent a subset of CD4+ T cells that are critically involved in balancing the reactivity of the immune system and preventing autoimmunity (40, 50). Alongside their role in preventing autoimmune reactions, Treg cells have been shown to control excessive inflammatory responses against pathogens (20, 48). On the other hand, a strict control of T effector (Teff) cell responses by Treg cells can promote pathogen persistence (9, 28, 44, 67). Some cell markers used to identify Treg cells are CD25, glucocorticoid-induced tumor necrosis factor receptor family-related gene (GITR), and the transcription factor forkhead box transcription factor P3 (Foxp3). In addition, the integrin αE(CD103)β7 is a marker for a subset of highly potent effector/memory-like Treg cells, and CD4+ CD103+ cells were found to be the most potent suppressors of inflammatory processes in disease models, such as colitis and arthritis (22, 23, 30, 54). Here, we used CD25, CD103, and Foxp3 to identify Treg cells. For the characterization of parasite-specific Teff cells, we used CD154, a marker recently shown to exhibit exquisite specificity for antigen-activated T cells (15, 26).

To investigate the roles of different CD4+ T-cell subsets during the chronic phase of a primary nematode infection, we used the gastrointestinal nematode H. polygyrus, which resides in the proximal third of the mouse duodenum for up to several months during primary infection. Mice become infected by ingestion of infective larvae (L3) that invade the duodenal wall, where development to the L4 stage takes place. L4 reenter the gut lumen and mature to adults, which are chronically maintained during primary infection (42). We used H. polygyrus infection to address the role of CD4+ Teff cells and Treg cells with regard to worm expulsion and the role of Treg cells in modulating Teff cell function. Our data demonstrate that adoptive transfer of Teff cells leads to protective immune responses in which antigen-specific CD4+ T cells produce predominantly IL-4 and IL-13. In contrast, CD4+ Treg cells from chronic infection show no effect on the adult worm burden after adoptive transfer, although they are highly potent suppressors in vitro.

MATERIALS AND METHODS

Mice, parasites, and infection.

H. polygyrus was maintained by serial passage in BALB/c mice. The infective larvae (L3) were obtained from feces culture, extensively washed, and stored in distilled water at 4°C. BALB/c and C57BL/6 mice were purchased from the Bundesinstitut für Risikobewertung, Berlin, Germany. C57BL/6-Tg(CAG-EGFP)C15-001-FJ001Osb mice were a gift of M. Okawa, Osaka University. Male mice at 7 to 10 weeks of age were infected with 200 L3 using a gavage tube. Infection was surveyed by fecal egg count. The worm burden was determined by collecting adult worms from the small intestine on the day of dissection. Animals were housed and handled following national guidelines and as approved by our animal ethics committee.

Preparation of adult worm antigen.

Soluble worm antigen was prepared from adult worms kept in culture in RPMI medium containing 100 U/ml penicillin and 100 μg/ml streptomycin for 24 h. The worm material was homogenized and sonicated (1 min; 60 W) on ice in phosphate-buffered saline (PBS) (pH 7.4). The homogenate was centrifuged (20 min; 20,000 × g; 4°C), and the supernatant was passed through a 0.4-μm filter (Schleicher & Schuell, Germany) for sterilization. The protein content was determined by the bicinchoninic acid test (Pierce). Antigen extracts were stored at −80°C until they were applied.

Antibodies, staining, and sorting reagents.

The following antibodies and secondary reagents were purchased from BD Biosciences (Heidelberg, Germany): αCD4 fluorescein isothiocyanate (FITC)/PerCP (RM4-5), αCD103 phycoerythrin (PE) (M290), αCD25 allophycocyanin (APC)/PerCP-Cy5.5 (PC61), αCD3e (145-2C11), αCD28 (37.51), αCD40 (HM40-3), αIL-10 PE (JES5-16E3), αIL-4 FITC (11B11), α-gamma interferon (αIFN-γ) FITC (XMG1.2), αCD8 PE (53-6.7), SA-PE-Cy7, and SA-APC. αCD103 biotin (M290), αCD19 FITC (ID3), αFcR II/III (2.4G2), and α-digoxigenin PE were kindly provided by the German Arthritis Research Center (Berlin, Germany). αCD154 (CD40L) APC (MR1) was obtained from Miltenyi Biotec (Bergisch-Gladbach, Germany). αIL-13 (38213.11) was obtained from R&D systems (Wiesbaden, Germany) and coupled with digoxigenin at the German Arthritis Research Center. Foxp3 staining was performed using the PE α-mouse Foxp3 staining set (clone FJK-16S) purchased from eBioscience (San Diego, CA). αCD4, αCD90, αPE, and αAPC microbeads, as well as FITC MultiSort kits, were obtained from Miltenyi Biotec.

Phenotypic analysis of lymphocytes by flow cytometry.

Mesenteric lymph node cells (MLNC) and splenocytes from naïve and H. polygyrus-infected mice were dissociated by passing organs through a steel mesh in PBS, pH 7.4, containing 0.2% bovine serum albumin. In some experiments, intraepithelial lymphocytes (IEL) and lamina propria lymphocytes (LPL) were isolated from the small intestine. After visible Peyer's patches were removed, the small intestine was opened longitudinally; washed in PBS, pH 7.4; and incubated in RPMI at 37°C, 150 rpm for 40 min. The small intestine was washed twice in PBS, and the supernatants were collected for isolation of IEL. The organ was cut into pieces and incubated (40 to 50 min; 37°C; 150 rpm) with collagenases VIII and D (40 μg/ml each; both from Sigma), and then tissue was removed with a mesh. The supernatants were spun, and the cells were layered on a column of Percoll (GE Healthcare, Uppsala, Sweden) with a 40%-70% gradient. The cells were spun at room temperature and 2,200 × g for 20 min, and then collected from the interphase, washed, and kept in PBS-0.2% bovine serum albumin. For detection of changes in lymphocyte composition, cell suspensions (1 × 106 total cells) were stained with αCD4, αCD8, and αCD19 monoclonal antibodies (MAbs). Treg cells were detected by staining them for CD4, CD25, and CD103. Nonspecific binding of the MAbs was blocked by the addition of αFcgRII/III (20 μg/ml). Intracellular detection of Foxp3 was performed according to the manufacturer's instructions. For intracellular detection of CD154 and cytokines, cells were fixed in PBS containing 2% formaldehyde for 15 min at room temperature. After permeabilization with 0.5% saponin (Sigma), the cells were blocked with whole rat IgG (0.1 mg/ml) for 15 min at 4°C to reduce nonspecific binding of MAbs and stained with αCD154 and two of the α-mouse cytokine MAbs for 30 min at 4°C. For combined detection of CD154 and Foxp3, CD154 was stained on the cell surface directly during in vitro stimulation in complete culture medium as described below (see “Culture conditions”). Cytometric analysis was performed using FACSCalibur or LSRII (BD Biosciences) and FlowJo software (Tree Star, Inc.).

Isolation of T-cell subsets.

The separation of T-cell subsets for transfers and in vitro stimulation was performed as follows. Cells were stained for CD25 (APC) and CD103 (PE). CD25+ and CD103+ cells were enriched by the AutoMACS magnetic separation system using αAPC and αPE magnetic beads. For isolation of the different regulatory subsets, the bead-positive fraction was stained with FITC-labeled αCD4, and the CD4+ CD25+ CD103+ cells and CD4+ CD25+ CD103 cell subsets were separated using a FACS Diva cell sorter (BD, Heidelberg, Germany). After complete removal of cells expressing CD25 and/or CD103, the negative fraction was used to isolate conventional CD4+ T cells using αCD4 beads. Naïve splenocytes depleted of T cells using αCD90 beads were irradiated (30 Gy) and used as antigen-presenting cells for in vitro assays. For some adoptive transfers, the whole CD4+ CD103+ subset (irrespective of CD25 expression) was isolated. Therefore, cells were stained for CD4 (FITC), CD25 (APC), and CD103 (PE). CD4+ cells were isolated using the FITC-MultiSort kit by AutoMACS. After removal of the beads, CD103+ cells were isolated from CD4+ cells using αPE beads.

Culture conditions.

Cell cultures were performed in cRPMI (BioChrom, Berlin, Germany) containing 10% fetal calf serum, 20 mM l-glutamine, 100 U/ml penicillin, and 100 μg/ml streptomycin as quadruplicates on 96-well plates. Culture of complete and Treg cell-depleted MLNC was performed with 3.5 × 105 cells for 72 h and a concentration of 12 μg/ml of adult worm antigen, and then the supernatants were harvested for cytokine detection. Culture of regulatory subsets and CD4+ CD25 CD103 responder cells for detection of polyclonal response to αCD3 stimulation (1 μg/ml) was performed with 2.5 × 104 CD4+ cells and 5 × 104 antigen-presenting cells per well. The cells were incubated for 48 h, followed by the addition of 1 μCi of [methyl-3H]thymidine (Amersham Pharmacia Biotech, Gent, Belgium) per well for 20 h to measure proliferation. In coincubation assays, the indicated numbers of Treg cells were added to naïve responder cells and antigen-presenting cells and treated as described above. In some assays, bone marrow-derived dendritic cells (DC) were used as antigen-presenting cells, together with sorted CD4+ T-cell populations. For the generation of DC, bone marrow was isolated from tibias and femurs of 6- to 8-week-old naïve BALB/c mice, and the cells were kept in 24-well plates at a concentration of 1.5 × 106/ml in cRPMI supplemented with 20 ng/ml of granulocyte-macrophage colony-stimulating factor (PeproTech, Hamburg, Germany) for 6 days, followed by incubation with 10 μg/ml of adult worm antigen for 12 h. Control cells were left untreated. DC (1 × 104) were cultured for 72 h with 5 × 104 T cells, and then the supernatants were harvested for cytokine enzyme-linked immunosorbent assay (ELISA) and the cells were snap-frozen in liquid nitrogen and stored at −80°C for real-time PCR. Recombinant mouse IL-2 (10 ng/ml; PeproTech, Hamburg, Germany) and 1 μg/ml αCD28 were added to some cultures for optimal stimulation. For measurement of CD154 (CD40L) expression of T cells and detection of cytokines in antigen-specifically activated cells, splenocytes and MLNC from infected animals were incubated in 24-well plates at a concentration of 4 × 107 cells per ml with 20 μg/ml of adult worm antigen and 1 μg/ml αCD28 for 12 h. To survey cytokine production, brefeldin A (5 μg/ml; Sigma) was added after the first 2 h of stimulation. After 12 h, the cells were washed and prepared for flow-cytometric analysis as described above. For surface staining of CD154, cells were incubated as indicated above but without the addition of brefeldin A. αCD154-APC, αFcR II/III MAbs (20 μg/ml), whole rat IgG (10 μg/ml), and αCD40 (to avoid rapid removal of CD154 from the cell surface after binding of CD40 expressed on antigen-presenting cells) were added to the culture. After 12 h, the cells were washed and stained for CD4, CD103, and Foxp3. We omitted staining of CD25 in these assays due to the unreliability of the marker with respect to Treg characterization after restimulation.

Histology.

Tissue samples from the proximal third of the small intestines of naïve and H. polygyrus-infected mice were fixed in 4% phosphate-buffered formalin, embedded in paraffin, and used for cross sections. Immunohistology for Foxp3-expressing cells was performed as described elsewhere (32, 33). Foxp3+ cells were counted in 10 high-power fields (40-fold magnification) randomly distributed in sections from each animal (Peyer's patch areas were excluded).

Cytokine analysis and quantitative PCR.

IL-4, IL-10, and IFN-γ in cell culture supernatants were quantified using OptEIA ELISA kits (BD Biosciences) according to the manufacturer's instructions. IL-13 and active transforming growth factor β1 (TGF-β1) were detected using DuoSets from R&D Systems. Active TGF-β1 was analyzed in culture supernatants without acidification. Transcript quantification by real-time PCR of IL-4 and IL-10 in distinct CD4+ Teff and Treg populations was performed after coincubation of T cells with naïve or H. polygyrus antigen-pretreated bone marrow-derived DC (see “Culture conditions” above). RNA extractions were performed using the RNeasy Mini Kit (Qiagen, Hilden, Germany), followed by digestion of DNA using the RNase-free DNase set (Qiagen) according to the manufacturer's instructions. RNA was reverse transcribed using the TaqMan reverse transcription reagent (Applied Biosystems, Warrington, United Kingdom) and oligo(dT)s. Quantitative real-time PCR was performed with the 7300 Real-Time PCR System (Applied Biosystems) using TaqMan reagents (Applied Biosystems). PCR amplifications were done in triplicates containing 3 μl of cDNA, 2 μl of 20× TaqMan-labeled primer mixture, and 10 μl of 2× TaqMan PCR buffer. The 20× TaqMan primer mixture consisted of two unlabeled PCR primers (900 nM [each] final concentration) and one 6-carboxyfluorescein dye-labeled TaqManMGB probe (250 nM final concentration). All primers were obtained from Applied Biosystems (IL-4 assay identifier, Mm00445259_m1; IL-10 assay identifier, Mm00439616_m1; GAPDH [glyceraldehyde-3-phosphate dehydrogenase] assay identifier, Mm99999915_g1). Real-time PCR was performed using the following conditions: 10 min of denaturation at 95°C, followed by 40 amplification cycles of 15 s at 95°C and 60 s at 60°C. The relative amounts of IL-10 and IL-4 mRNA were normalized to the endogenous reference GAPDH. Quantification of transcripts in cells cultured in the presence of DC pretreated with H. polygyrus antigen was done relative to cells cultured with naïve DC using the 2−ΔΔCT method as described elsewhere (31).

Adoptive-transfer experiments.

Sorted CD4+ T-cell subpopulations (5 × 105 cells per animal) were injected intraperitoneally into naïve mice in 0.2 ml of sterile PBS. Control animals received PBS only. One day after transfer, the mice were infected with approximately 200 L3 larvae. Four weeks after infection, animals were sacrificed, and the number of adult worms in each animal was determined and calculated as a percentage of the exact dose of applied L3 (set as 100%). The success of the infection was determined by surveying the fecal egg output starting on day 10 postinfection (p.i.). To survey cell survival and to trace transferred cells, the mice received 1 × 107 carboxyfluoroscein succinimidyl ester (CFSE)-labeled or enhanced green fluorescent protein (EGFP)-expressing CD4+ cells. Reanalysis by flow cytometry was performed 6 days after transfer to the spleen, MLN, and small intestine.

Statistical analysis.

Statistical analysis was performed using GraphPad Prism software (San Diego, CA). Statistical significance as indicated in the figure legends was analyzed by either the Mann-Whitney test or analysis of variance (ANOVA), in combination with Bonferroni posttests.

RESULTS

Analysis of lymphocytes in MLNs of mice chronically infected with H. polygyrus.

To gather information about changes occurring in the MLN after infection with H. polygyrus, we analyzed the lymphocyte composition of the MLN in the chronic phase of infection (28 days p.i.). The absolute cell numbers obtained from the MLN showed a 5.2-fold increase (P < 0.05) in infected compared to naïve animals (Table 1). CD4+ and CD8+ T cells, as well as B cells, were analyzed by flow cytometry staining. We found a 4.7-fold (P < 0.05) increase in absolute CD4+ and a 4.3-fold (P < 0.05) increase in absolute CD8+ T-cell numbers. The strongest increase in total numbers, however, was found for B cells (8.3-fold; P < 0.05) (Table 1). Interestingly, the number of CD25+ CD103+ T cells increased 7.8-fold (P < 0.05) within the CD4+ T-cell compartment, surpassing the outgrowth of CD25+ CD103 T cells (5.1-fold; P < 0.05) and CD25 CD103 Teff cells (4.5-fold; P < 0.05) (Table 1).

TABLE 1.

Total MLNC numbers and lymphocyte composition of MLNs from naïve and H. polygyrus-infected animals

Cell type No.
Increase (fold)
Naïve 28 days p.i.
CD4+ 9.32 × 106 ± 1.40 × 106b 43.54 × 106 ± 5.17 × 106b,c 4.7
    CD25 CD103a 8.28 × 106 ± 1.21 × 106 36.93 × 106 ± 4.35 × 106c 4.5
    CD25+ CD103a 0.79 × 106 ± 0.12 × 106 4.06 × 106 ± 0.29 × 106c 5.1
    CD25+ CD103+a 0.21 × 106 ± 0.06 × 106 1.64 × 106 ± 0.28 × 106c 7.8
CD8+ 2.89 × 106 ± 1.01 × 106 12.71 × 106 ± 3.19 × 106c 4.3
B cells (CD19+) 4.67 × 106 ± 2.12 × 106 38.99 × 106 ± 15.54 × 106c 8.3
Total 1.72 × 107 ± 0.29 × 107 8.96 × 107 ± 1.1 × 107c 5.2
a

CD4+ T-cell subset.

b

Mean ± standard error of the mean of four (naïve) and five (28-day p.i.) animals. Similar data were obtained in four experiments.

c

Statistical significance for comparison of naïve to infected animals as determined by the Mann-Whitney test (P < 0.05).

Analysis of Treg cells during the course of infection.

To further characterize the regulatory CD4+ T-cell compartment, we analyzed the frequency of Treg cells in the MLNs and spleens of H. polygyrus-infected mice at different time points after infection. Flow cytometry analysis was performed after staining of CD4, CD25, CD103, and the transcription factor Foxp3. By detecting CD25 and CD103, we were able to distinguish between CD4+ CD25+ CD103 naturally occurring and CD4+ CD25+ CD103+ effector/memory-like Treg cells (Fig. 1A). Foxp3 expression levels were >90% in CD4+ CD25+ CD103 cells at most time points analyzed (naïve mice and 3, 21, and 28 days p.i.), with the exception of 6 days p.i., showing a significant decrease in Foxp3+ cells within this compartment (naïve, 95.75% ± 0.05%; 6 days p.i., 86.72% ± 1.11%; P < 0.03), arguing for an increased proportion of recently activated effector cells present in MLNC at this time point (Fig. 1B). The CD4+ CD25+ CD103+ subset displayed Foxp3 expression levels of >96% at all time points analyzed (Fig. 1B and not shown), while few (<1.8%) CD4+ CD25 CD103 cells expressed Foxp3 (not shown). Comparing percentages of CD4+ Foxp3+ cells between naïve and infected animals (3, 6, 21, and 28 days p.i.) revealed no significant differences (Fig. 1E and not shown). Thus, the increase in total Treg cells during infection reflects the increase in CD4+ cell numbers. However, we detected significant changes concerning the frequency of effector/memory-like Treg cells after infection with H. polygyrus. Within the MLN draining the site of infection, we determined a significant increase (P < 0.03) in CD103+ Treg percentages as early as day 6 days p.i. in comparison to naïve animals (Fig. 1C). Within the spleen, a significantly increased percentage was detected from day 12 p.i. onward (P < 0.03) (Fig. 1D). The higher frequency of Treg cells expressing CD103 was stable in both lymphoid compartments until the chronic phase of infection (Fig. 1C and D) and returned to the level of naïve mice when adult worms were expelled after 8 to 12 weeks (data not shown). The level of CD25+ CD103 Treg cells showed no such increase (data not shown). When analyzing the ratio of CD103+ and CD103 Treg cells after gating on all Foxp+ cells, we rather determined a decrease in CD103 cells in infected animals at the chronic stage, counterbalancing the increase in CD103+ cells (Fig. 1F). This finding argues for a conversion of naturally occurring Treg cells into effector/memory-like Treg cells, probably directly related to the infection.

FIG. 1.

FIG. 1.

Comparison of Treg numbers in naive and H. polygyrus (H.p.)-infected mice. (A) Treg cells were detected by flow cytometry based on surface expression of CD4, CD25, and CD103. Shown are plots derived from CD4+ MLNC of naïve and chronically infected animals. The data are representative of four animals per group and four independent experiments. (B) The predominantly regulatory phenotype of CD4+ CD25+ CD103 (top) and CD4+ CD25+ CD103+ (bottom) cells derived from MLNs could be confirmed by intracellular staining of Foxp3. Four animals per group were analyzed in two independent experiments. (C and D) The frequencies of CD25+ CD103+ cells within CD4+ lymphocytes as detected in MLNs (C) and spleens (D) of animals at different time points after infection (black bars) compared to naïve controls (open bars). The data were derived from four animals per group, and analysis of Treg cell numbers was performed at least twice for each time point. Means plus standard errors of the mean (SEM) are shown. (E) Frequencies of total Foxp3+ cells within CD4+ lymphocytes from MLN. Means plus SEM of four animals per group are shown. The data are representative of two independent experiments. (F) Proportion of CD25+ CD103 and CD25+ CD103+ cells in CD4+ Foxp3+ cells. Means plus SEM of four animals per group are shown. The data are representative of two independent experiments. (G) Cross sections of the proximal third of the small intestine were stained for Foxp3-expressing cells. Foxp3+ Treg cells were found in the epithelium and in the lamina propria (depicted by black arrowheads). The white arrowheads indicate tissue-dwelling larvae of H. polygyrus at day 6 p.i. (H) Significant increases in Treg cells within the intestine were detected on days 3, 6, and 12 p.i. Stained cells in 10 high-power fields (HPF) (40-fold magnification) per animal were counted. Means of single animals (closed circles) and means of groups (horizontal lines) are shown. *, significant difference between naïve and infected animals as determined by the Mann-Whitney test (P < 0.05).

To test whether the changes in Treg numbers found in the lymphatic organs were also reflected at the site of inflammation, we detected Foxp3-expressing cells in sections of the proximal third of the small intestine (Fig. 1G). In clear contrast to the findings in lymphatic organs, we detected a transient maximal accumulation of Foxp3+ cells at day 6 p.i. (P < 0.016), which gradually returned to the basal level seen in naïve animals at later time points of infection (Fig. 1H). The kinetics of Foxp3+ cells was mirrored by a maximal production of IL-10 and TGF-β1 in the small-intestinal tissue at day 6 p.i. (data not shown). Taken together, these data show that Treg cells with an effector/memory-like phenotype increase strongly during infection with H. polygyrus in the MLN and spleen, while Foxp3-expressing cells accumulate only transiently at the site of infection.

Cytokine production of complete and CD25/CD103-depleted MLNC.

MLNC of H. polygyrus-infected mice were isolated at different time points after infection and cultured in the presence of H. polygyrus antigen to analyze their cytokine profile. Our data show that the key Th2 cytokines IL-4 and IL-13 in particular, as well as IL-10, were readily produced in high concentrations during the early phase of infection (6 days p.i.), accompanied by a weak IFN-γ response (Fig. 2A to D). Similar amounts of cytokines were found during the acute phase (12 days p.i.), except for IFN-γ, which was only marginally produced at this time point (Fig. 2A). At the chronic phase of infection (28 days p.i.), all cytokines analyzed were produced in smaller amounts, especially IL-13 and IL-10. Although it did not reach statistical significance, the trend toward lower cytokine production by MLNC from the chronic phase argues for a generally down-regulated parasite-specific response. Active TGF-β1 was detected only in very small amounts (<15 pg/ml) irrespective of the time point (data not shown). Interestingly, depletion of cells with a mainly regulatory phenotype (carrying the surface markers CD25 and CD103) (Fig. 2E) resulted in drastic changes in cytokine production of the remaining MLNC (Fig. 2A to D). First, in the early phase of infection (6 days p.i.), all cytokines analyzed were produced in smaller amounts after depletion of CD25+ and CD103+ cells. The finding of lower IL-4, IL-13, IL-10, TGF-β1 (not shown), and IFN-γ levels in cultures depleted of cells carrying CD25 and/or CD103 argues for depletion of not only Treg cells, but also recently activated Teff cells at this early time point. We could confirm the presence of effector cells within the CD4+ CD25+ CD103 subset at 6 days p.i. by detection of Foxp3 expression (showing a decline at 6 days p.i.) (Fig. 1B) and by analyzing IL-4 and IL-10 mRNA levels in different CD4+ T-cell subsets kept in cocultures with DC presenting H. polygyrus antigens (Fig. 2F). We determined that CD4+ CD25+ CD103 T cells expressed large amounts of IL-4 and IL-10 transcripts at this early time point, indicating the presence of Teff and Treg cells within this population. In contrast, the CD4+ CD25+ CD103+ Treg population from the early infection exclusively expressed high levels of IL-10 (Fig. 2F). As we did not analyze the segregation of the IL-10 production to Foxp+ and Foxp3 cells within the CD4+ CD25+ CD103 compartment, we cannot exclude a contribution of recently activated effector cells to the IL-10 production at the early time point. Hence, the depletion of cells expressing CD25 and/or CD103 at the early time point removed a significant proportion of recently activated Teff cells, as well as Treg cells, as seen by diminished production of the Th2 cytokines IL-4, IL-13, and IL-10 by the remaining cells. The depletion had no effect with respect to IL-10 production at later time points (Fig. 2D), arguing that cells other than Treg cells may also represent important IL-10 sources during the acute and chronic phases. Interestingly, the IL-4 response in the chronic phase of infection was more vigorous after depletion, indicating a suppressive effect of Treg cells on the Th2 response in the chronic phase of infection (Fig. 2B).

FIG. 2.

FIG. 2.

Cytokine production of MLNC during H. polygyrus infection. (A to D) The cytokine response of MLNC after restimulation with H. polygyrus adult worm antigen was analyzed with cells from animals at days 6, 12, and 28 p.i. The cytokine response of complete MLNC (open bars) was compared to that of MLNC depleted of cells expressing CD25 and CD103 (hatched bars). IFN-γ (A), IL-4 (B), IL-13 (C), and IL-10 (D) production was analyzed. The means plus standard deviations (SD) of three cell pools of two animals each are shown. *, statistical significance comparing cytokine responses before and after depletion as determined by two-way ANOVA, followed by Bonferroni posttests (P < 0.05). The data are representative of two independent experiments. (E) The efficiency of Treg depletion was determined by flow cytometry. The expression of CD25 and CD103 (upper row) and Foxp3 (lower row) in CD4+ cells before (left) and after (right) depletion is shown. (F) The relative expression of IL-4 and IL-10 mRNAs by CD4+ T-cell subsets after coincubation with H. polygyrus-primed DC was assessed at day 6 p.i. Means plus SD of three measurements are shown.

Adoptive transfer of CD4+ T cells from H. polygyrus-infected mice.

To analyze the functions of different CD4+ T-cell subsets, we performed adoptive transfers of CD4+ Teff cells in comparison to CD4+ Treg subsets derived from chronically H. polygyrus-infected mice (28 days p.i.). Teff cells (CD4+ CD25 CD103) and CD4+ T cells with a regulatory phenotype, namely, CD4+ CD25+ CD103 and CD4+ CD25+ CD103+ T cells, were transferred to naïve recipients. Control animals received PBS only. One day after transfer, the recipients were infected with a defined dose of larvae. The purity of Teff cells was >98% for expression of CD4+, with <3% remaining CD25+ and/or CD103+ cells, whereas the Treg compartments were >90% CD4+ CD25+ CD103 cells and >89% CD4+ CD25+ CD103+ cells, respectively (Fig. 3A). Four weeks after infection, the adult worm burdens of recipients and control animals were assessed. We detected a significant reduction of 43.7% (P < 0.008) in the worm burden in animals receiving Teff cells (CD4+ CD25 CD103) compared to the PBS control group (Fig. 3B). In contrast, transfer of CD4+ CD25 CD103 T cells from naive mice had no influence (data not shown), indicating that the reduction was due to parasite-specific Teff cells. The protective role could be solely ascribed to CD4+ T cells, as the transfer of CD4 cells had no effect (data not shown). Animals receiving CD4+ CD25+ CD103 Treg cells (comprising >90% Foxp3+ cells) had worm burdens comparable to that of the PBS control. Similarly, the transfer of CD4+ CD25+ CD103+ Treg cells (comprising >95% Foxp3+ cells) did not result in significant changes. In contrast, transfer of a heterogeneous T-cell population containing Treg and Teff cells, namely, CD4+ CD103+ T cells (with a purity of >95% CD4+ cells and >85% CD103+ cells but comprising only about 70% Foxp3+ cells) led to a significant decrease in worm numbers (P < 0.03) comparable to pure CD4+ Teff cells (Fig. 3B). We verified cell survival in separate experiments using CFSE-labeled CD4+ cells (data not shown). In addition, we performed transfers with EGFP-expressing CD4+ cells from chronically infected donors. The recipients were infected the following day. Six days after transfer, spleen and MLN, as well as IEL and LPL from the small intestine, were analyzed for transferred cells (Fig. 3C and D). We were able to trace EGFP+ cells in all analyzed organs with no significant accumulation in any of the compartments. The slightly elevated numbers of EGFP+ cells within MLNC compared to splenocytes merely reflects the higher CD4+ T-cell numbers found in lymph nodes (Fig. 3D). We also analyzed percentages of Treg cells according to expression of CD25, CD103, and Foxp3 within the EGFP+ population and could show that Treg cells survived the transfer (data not shown).

FIG. 3.

FIG. 3.

Influence of adoptive CD4+ T-cell transfer on adult worm burden. T cells of the indicated subtypes (5 × 105) obtained from MLNs and spleens of chronically H. polygyrus-infected mice were transferred to recipients that were subsequently infected with H. polygyrus larvae. Control animals received PBS only. (A) Purity of transferred cells as determined by flow cytometry. Representative data from one of three independent experiments are shown. (B) Adult worm burden in recipients 28 days p.i. Worm counts are shown as percentages of the number of applied larvae. Group sizes varied between 5 and 20 animals. The data originated from three individual experiments. Individual worm counts and medians are shown. The asterisks show statistical significance as determined by a Kruskal-Wallis test followed by a Mann-Whitney test: *, P < 0.05; **, P < 0.01. (C) Tracing of transferred cells in C57BL/6 mice receiving 1 × 107 CD4+ cells from chronically infected EGFP-expressing donors. The recipients were infected with H. polygyrus the following day. Examples of flow cytometry plots derived from splenocytes, MLNC, and small-intestinal IEL and LPL 6 days after transfer are shown. (D) Percentages of EGFP+ cells within lymphocytes of recipients. Means plus standard deviations for four animals are shown. The data are representative of two experiments.

Hence, CD4+ Teff cells from the chronic phase of an H. polygyrus infection transferred protection to naïve recipients in contrast to naturally occurring or effector/memory-like Treg cells, which had no intrinsic effect on worm development. Furthermore, transfer of a mixed T-cell population comprising effector and regulatory T cells showed that Treg cells were not efficient in suppressing the protective effect mediated by Teff cells.

Cytokine production of CD4+ T-cell subsets in the chronic phase of infection.

We then analyzed the cytokine production of the CD4+ T-cell subsets used for the transfer experiments to gain information about mediators involved in worm expulsion or establishment. H. polygyrus-treated DC were used as antigen-presenting cells to stimulate the T-cell subsets. We determined that Teff cells (CD4+ CD25 CD103) released IL-4 and IL-13 in the presence of H. polygyrus-primed DC in contrast to Treg cells (CD4+ CD25+ CD103 or CD4+ CD25+ CD103+) (Fig. 4A and B). However, CD4+ CD25+ CD103+ T cells, in particular, released significant amounts of IL-10, arguing for the effector/memory-like Treg cell subset as an important source of IL-10 (Fig. 4C). Incubation of the CD4+ subsets with naïve DC resulted in marginal release of cytokines by T cells, clearly showing the antigen specificity of the cytokine response. Analysis of active TGF-β1 in culture supernatants revealed only marginal increases in cultures with antigen-loaded DC compared to those with naïve DC (data not shown). The generally lower cytokine levels in cultures of separated Teff cells compared to complete MLNC cultures (Fig. 2) is probably due to fewer CD4+ T cells present in DC cocultures than in preparations of whole MLNC and to other cytokine-producing cells present in MLNC cultures, like basophils, mast cells, and eosinophils (16).

FIG. 4.

FIG. 4.

Cytokine production by CD4+ T-cell subsets. CD4+ T cells were isolated from pooled MLNC and splenocytes of eight mice in the chronic phase of infection (28 days p.i.) according to the indicated surface marker expression. T-cell subsets were incubated for 72 h with naïve bone marrow-derived DC (nDC) or DC pretreated with H. polygyrus adult worm antigen (HpDC). (A to C) Release of IL-4 (A), IL-13 (B), and IL-10 (C) was detected by ELISA. Means plus standard deviations (SD) of triplicate determinations are shown, and the data are representative of three independent experiments. (D to F) Release of IL-4 (D), IL-13 (E), and IL-10 (F) after addition of recombinant mouse IL-2 and αCD28 to cultures. Mean values plus SD of triplicate determinations of one of two independent experiments with similar results are shown.

While only CD4+ CD25+ CD103+ T cells produced IL-10 in an antigen-specific manner, we found that both CD25-expressing T-cell subsets produced large amounts of IL-10 when optimized conditions were provided by addition of exogenous IL-2 and enhanced costimulation through αCD28 antibodies (Fig. 4F). Similarly, IL-4 and IL-13 were produced in much larger amounts almost exclusively by CD4+ CD25 CD103 Teff cells after enhanced costimulation was provided (Fig. 4D and E). Levels of active TGF-β1 were only marginally affected (data not shown). None of the CD4+ T-cell subsets from H. polygyrus-infected mice was found to produce relevant amounts of the Th1 cytokine IFN-γ (data not shown). Hence, our data indicate a dominant and parasite-specific Th2 response by CD4+ effector cells from the chronic phase of infection, while effector/memory-like CD4+ CD25+ CD103+ Treg cells probably produce IL-10 in response to H. polygyrus antigen.

Distribution of antigen-specific CD4+ T cells and their cytokine production.

To assess the distribution of H. polygyrus-specific CD4+ T cells within the Teff and Treg cell populations, we examined the expression of CD154 as a marker of antigen-specific CD4+ T-cell activation (15, 26). As the detection of CD154 directly ex vivo is not possible, we used an optimized in vitro protocol (26). Comparison of CD4+ T cells from spleen and MLN showed that in both sites, about 0.2% (MLN, 0.233 ± 0.036 naïve versus infected [P < 0.001]; spleen, 0.246 ± 0.040 naïve versus infected [P < 0.0003]) of CD4+ CD103 Teff cells expressed CD154 when restimulated with adult worm antigen in vitro (Fig. 5A and B). By additional detection of CD103 and Foxp3, we were able to distinguish between CD154 expression on Teff and Treg cells.

FIG. 5.

FIG. 5.

Distribution of antigen-specific CD4+ T cells (CD154+) and their cytokine responses. (A) Shown are examples of fluorescence-activated cell sorter plots of MLNC from naïve mice (upper row) and H. polygyrus-infected mice at day 28 p.i. (lower row). The cells were restimulated with H. polygyrus antigen in vitro and stained for CD4, CD154, CD103, and Foxp3. CD4+ cells were plotted for expression of CD103 and Foxp3 (left). The middle plots show CD154 expression within the CD4+ CD103+ Foxp3 effector population. The right-hand plots show CD154 expression by CD4+ CD103 Foxp3 effectors. (B and C) Frequencies of CD154-expressing cells within the CD4+ CD103 Foxp3 (B) and CD4+ CD103+ Foxp3 (C) effector populations of MLNs (black bars) and spleens (open bars) after restimulation in vitro. The data were obtained from eight infected and seven naïve mice. Means plus standard errors of the mean (SEM) are shown. The data are representative of three independent experiments. (D) Intracellular staining of cytokines within restimulated CD4+ T cells from spleens (28 days p.i.). The cells were gated for expression of CD4 and CD154 (left), and IL-4 and IL-13 (upper row) or IFN-γ and IL-10 (lower row) were determined in CD4+ CD154 cells (center) and CD4+ CD154+ cells (right). The cytometry plots shown are representative of a group of eight infected mice. (E) IL-4/IL-13 and IFN-γ/IL-10 (F) responses in CD4+ CD154+ cells after restimulation with H. polygyrus antigen. Means plus SEM of eight animals are shown. The asterisks show statistical significance comparing cells from naïve and infected animals as determined by the Mann-Whitney test: **, P < 0.01; ***, P < 0.001.

Interestingly, when analyzing the level of CD154 on CD103+ cells not coexpressing Foxp3 and therefore not of regulatory phenotype, we found clearly higher percentages of antigen-specific cells within this Teff subpopulation in MLNs (1.701% ± 0.306% CD4+ CD103 versus CD4+ CD103+; P < 0.0006) and spleens (0.586% ± 0.144% CD4+ CD103 versus CD4+ CD103+; P < 0.01) (Fig. 5A and C) of infected animals in comparison to noninfected mice. In contrast, only low levels of CD154 expression were detected on Foxp3+ Treg cells from MLN (<0.03%) and spleen (<0.05%) (data not shown). These data indicate that mice that received T cells sorted for coexpression of CD4 and CD103 (irrespective of CD25 expression) in transfer studies (Fig. 3B) received a highly enriched Treg population that still contained a small number of antigen-specific effector cells not expressing Foxp3. The percentage of antigen-specific effectors within the transferred CD4+ CD103+ cell compartment was as low as 0.2%, according to CD154 expression, comparable to the values for antigen-specific cells within the CD4+ CD103 effector compartment. These CD103+ effectors may have led to protection, even though a large number of Treg cells was cotransferred, arguing for an insufficient capacity of the Treg cells to control the antigen-specific effectors.

We next determined the cytokine production of the antigen-specific CD154+ CD4+ T cells. Investigation of IL-4, IL-13, IL-10, and IFN-γ production after in vitro restimulation (Fig. 5D) revealed a dominant Th2 response in CD154+ CD4+ T cells of chronically infected mice, characterized by high expression levels of IL-4 (24.55% ± 1.32%), IL-13 (10.48% ± 1.99%), or both (10.26% ± 1.02%) (Fig. 5D and E). Only low levels of IFN-γ-producing cells were detected in the antigen-specific T-cell compartment (4.47% ± 0.82%), and the frequencies of IL-10-producing cells were hardly distinguishable from nonspecific background (1.60% ± 0.37%) (Fig. 5D and F). These data again indicate that the lower worm burden in recipients receiving CD103-expressing CD4+ T cells might be due to cotransfer of antigen-specific effector cells that were able to produce IL-4 and IL-13, thereby mediating worm expulsion.

Suppressive effect of Treg cells from H. polygyrus-infected mice in vitro.

To further investigate Treg cells from worm-infected animals, we analyzed the suppressive activities of sorted Treg subsets in vitro. CD4+ CD25+ CD103+ and CD4+ CD25+ CD103 Treg cells were isolated from naïve and worm-infected mice in the acute (12 days p.i.) and chronic (28 days p.i.) phases of infection. The cells were added to Treg-depleted CD4+ T cells from naïve mice, which were stimulated polyclonally by αCD3 antibodies. CD4+ CD25+ CD103+ Treg cells from infected mice suppressed the proliferation of naïve responder CD4+ T cells more vigorously than their counterparts from naïve controls (Fig. 6A and Table 2). For the lowest ratio of CD4+ CD25+ CD103+ Treg cells to responder cells (1:20), reflecting the in vivo situation, we found that Treg cells from the chronic phase of infection exhibited the highest suppressive efficiency (P < 0.001 compared to naïve cells and day 12 p.i.). The CD4+ CD25+ CD103+ Treg cells derived from infected animals were more efficient in mediating suppression than CD4+ CD25+ CD103 Treg cells (P < 0.03 for all tested ratios) (Fig. 6B). As expected, both Treg subsets showed an anergic phenotype after αCD3 stimulation (data not shown). These data clearly indicate the high in vitro suppressive capacity of CD4+ CD25+ CD103+ Treg cells derived from the chronic phase of infection with regard to activation and proliferation of CD4+ T cells.

FIG. 6.

FIG. 6.

Suppressive capacities of Treg cells in vitro. CD4+ CD25+ CD103 and CD4+ CD25+ CD103+ Treg cells were purified from splenocytes and MLNC of H. polygyrus-infected (12 and 28 days p.i.) and naïve mice. CD4+ CD25 CD103 responder cells were isolated from naïve animals. (A and B) The suppressive capacities of CD4+ CD25+ CD103+ Treg cells (A) and of CD4+ CD25+ CD103 Treg cells (B) from naïve mice (open bars) and infected animals at the acute phase (12 days p.i.) (gray bars) or chronic phase (28 days p.i.) (black bars) of infection were analyzed. The ratios of Treg cells and responder CD4+ T cells are indicated. Proliferation of CD4+ T cells after stimulation with αCD3 antibodies was detected by [3H]thymidine uptake. Means plus standard errors of the mean of quintuple determinations are shown. The data shown are representative of two independent experiments.

TABLE 2.

Statistical analysis of suppressive efficiencies of Treg cell subsets from naive and H. polygyrus-infected mice

Treg/CD4Respa ratio CD4+ CD25+ CD103b
CD4+ CD25+ CD103+b
Naïve vs. 12 days p.i. Naïve vs. 28 days p.i. 12 vs. 28 days p.i. Naïve vs. 12 days p.i. Naïve vs. 28 days p.i. 12 vs. 28 days p.i.
1:20 NS P < 0.001 P < 0.01 P < 0.001 P < 0.001 P < 0.001
1:10 P < 0.001 P < 0.001 NS P < 0.001 P < 0.001 NS
1:5 NS P < 0.01 NS P < 0.001 P < 0.001 NS
1:2 P < 0.001 NS P < 0.05 NS NS NS
1:1 NS NS NS NS NS NS
a

Ratio of Treg cells to naïve CD4+ responders.

b

Treg cell subset analyzed for suppressive efficiency. Statistical significance as determined by two-way ANOVA, followed by a Bonferroni posttest. NS, not significant.

DISCUSSION

In this study, we investigated the roles of different CD4+ T-cell subsets in a chronic intestinal worm infection. Adoptive transfer of CD4+ cells from the chronic phase of an H. polygyrus infection containing small numbers of antigen-specific Teff cells led to significant protection in recipients. In contrast, adoptive transfer of Treg cells did not alter the worm burden, although Treg cells significantly suppressed CD4+ T-cell proliferation in vitro. Hence, our data clearly point to the importance of CD4+ Teff cells in mediating host protection and worm expulsion. Our data provide evidence for the following traits of Teff/Treg interaction. First, Teff cells that arise during a primary H. polygyrus infection exhibit the ability to mediate protection. Second, Teff cells persist in chronically infected mice, despite strong cellular suppression (10, 49, 52). Third, about 0.24% of CD4+ Teff cells are antigen-specific cells, based on expression of CD154, containing highly enriched frequencies of IL-4- and IL-13-producing cells responding to parasite antigens. Fourth, Treg cells are probably an important source of IL-10 during chronic infection. Fifth, elevated numbers of effector/memory-like Treg cells persist in lymphatic organs until the chronic phase, and Treg cells derived from infected animals show an augmented suppressive capacity in vitro.

Our study revealed a significant and permanent increase in effector/memory-like (CD103+) Treg cell numbers in lymphatic organs as early as day 6 p.i. However, only a transient increase in Treg cells was detected at the site of inflammation, which might represent a host reaction to control inflammatory responses induced by innate immune and Teff cells. The peak of Treg cells in the small intestine at day 6 p.i. is in accordance with the time point of intense inflammation around tissue-invading larvae, characterized by accumulation of mainly granulocytes and, to a lesser extent, macrophages and CD4+ T cells (4, 43). Interestingly, it has been shown that Treg cells not only control inflammatory responses directly or indirectly driven by Teff cells, but also suppress innate immune responses in the absence of Teff cells (36). Therefore, it is conceivable that Treg cells induced by the nematodes are recruited to modulate innate and adaptive immune responses. The resulting immunosuppression could control excessive pathology and favor prolonged parasite survival. Helminth-induced Treg cells have been shown to be involved in immunomodulation in various helminth infections, such as human infections with Onchocerca volvulus (51) or murine infections with Litomosoides sigmodontis (59), H. polygyrus (14), and Schistosoma mansoni (6, 29, 37). The general notion is that during helminth infections, Treg cells might have the function of facilitating parasite survival and diminishing immunopathology (34, 35), but their concrete role is not yet fully elucidated.

We provide evidence that during the early phase of infection, activated Teff and Treg cells are both important sources of IL-10. However, during the chronic phase of infection, Treg cells probably represent the main T-cell source of IL-10. Hence, our data indicate that not only Treg cells, but also effector cells provide IL-10 that could have a role in dampening exaggerated inflammation, as shown previously for Th1 and Th2 effector cells (3, 8, 25, 61). Whether Treg cells in nematode infections mediate suppression via IL-10, TGF-β, or both is still under investigation (8, 27, 39, 59). Increases in TGF-β1 expression by CD4+ and CD4 cells and surface-bound TGF-β1 on CD4+ cells, as well as increased plasma levels of active TGF-β1 during infection with H. polygyrus, have been shown by others (14, 56, 68). In our system, Treg cells from the chronic stage of infection seem to exert their effects via IL-10, supported by the fact that we detected only low levels of active Treg-derived TGF-β1. Of note, our analysis of the TGF-β1 production by CD4+ T-cell subsets is restricted to the secreted active form, and currently we cannot exclude differences with respect to the secreted inactive form of the molecule. By analyzing tissue samples from the small intestine, we found elevated TGF-β1 levels at the site of infection in the early phase (6 days p.i.) (data not shown), perhaps indicating tissue repair in this phase of active inflammation.

In addition to T cells, B cells can also serve as major sources of IL-10 (45, 46), and B cells in protozoan and helminth infections have been shown to produce IL-10 in response to pathogen-derived antigens (2, 17, 46). To date, few data on B cells as a source of IL-10 in intestinal nematode infections are available. We detected large increases in B-cell numbers in MLNs of infected animals, and further analysis of these cells as possible source of IL-10 during H. polygyrus infection is needed.

This study showed that especially CD4+ CD25+ CD103+ T cells from mice chronically infected with H. polygyrus produce IL-10 in a parasite-specific manner. The high frequency of Foxp3-expressing cells within this T-cell population strongly suggests effector/memory-like Treg cells as a parasite-specific source for IL-10, although we cannot completely rule out the possibility that Foxp3 effector cells or adaptive Treg cells may contribute to the detected IL-10 production. In mice infected with S. mansoni, CD4+ CD25+ T cells, whether Foxp3+ or Foxp3, have been identified as sources of IL-10 (6, 58).

We revealed a disproportionate increase in CD25+ CD103+ Treg cells within the CD4+ T-cell compartments of infected animals. Similar to what has been described for infection with S. mansoni (6), we detected only minor changes in the total Treg cell numbers as determined from Foxp3 expression in lymphatic organs. However, we found a significant increase in cells expressing CD103 within the regulatory T-cell compartment, arguing for a specific role of these effector/memory-like Treg cells in modulating the immune response to H. polygyrus. To test this hypothesis, we performed suppression assays with responder CD4+ T cells from naïve mice. These in vitro assays demonstrated that especially CD4+ CD25+ CD103+ Treg cells isolated from the chronic phase of infection strongly suppressed the proliferation of CD4+ T cells when added in a physiological ratio of Treg to responder cells (1:20). A particularly potent suppressive capability of CD4+ CD25+ CD103+ Treg cells in comparison to CD4+ CD25+ CD103 Treg cells in vitro and in vivo has also been shown by others (30, 57). We expected that adoptive transfer of Treg cells might interfere with the primary Th2 response to H. polygyrus, entailing increased worm burdens, as studies on protective immunity against H. polygyrus have shown that severe combined immunodeficient mice lacking T and B cells and mice depleted of CD4+ T cells harbor higher worm burdens than wild-type or untreated controls (63, 64). However, in spite of the high suppressive capacity of CD103+ Treg cells in vitro, our adoptive-transfer model did not reveal an influence of Treg cells on the worm burden in vivo. Transfer of CD4+ CD25+ CD103+ Treg cells, as well as CD4+ CD25+ CD103 Treg cells, did not alter worm numbers, whereas transfer of a mixture of Treg and Teff cells (CD4+ CD103+ T cells) significantly lowered worm burdens, arguing against a major control of effector responses by Treg cells in vivo. The discrepancy between the suppressive activity of CD103+ Treg cells in vitro and the failure to interfere with worm expulsion in vivo might be due to multiple factors. First, Treg cells might be involved in suppressing pathology, but not in facilitating worm survival. Second, the transferred CD4+ CD103+ T-cell population represented a heterogeneous pool of Treg and Teff cells containing the lowest percentage of Foxp3+ cells (<70%) in comparison to the CD4+ CD25+ CD103+ and CD4+ CD25+ CD103 subsets (both ≥90%). Third, reduction of the adult worm burden in recipients receiving CD4+ CD103+ T cells coincided with a significant proportion of parasite-specific effector cells within the CD103+ Foxp3 cell compartment, as shown by expression of CD154 after antigen stimulation. Finally, the conditions in vitro versus in vivo were clearly different. In vitro, Treg cells drastically inhibited the proliferation of naïve CD4+ responder T cells after polyclonal stimulation, whereas in vivo, Treg cells had to combat antigen-specifically activated Teff cells.

We have provided data on the distribution and cytokine profile of parasite-specific CD4+ cells using the marker CD154, recently shown to exhibit exquisite specificity for antigen-activated CD4+ cells in the human and mouse systems (15, 26). Our approach revealed low percentages (<1%) of antigen-specific cells in the chronic phase, producing predominantly IL-4 and IL-13. Although cytokine-producing CD4+ T cells were also detected at low percentages among CD154 cells, the responders to worm antigens were highly enriched within the CD154+ population. In contrast, much higher frequencies of IL-4-producing cells are described for infections with H. polygyrus in GFP-IL-4 reporter mice (41). Several factors may have led to the marked discrepancies. First, Mohrs et al. reported that not all GFP+ cells secrete IL-4 in response to stimulation with H. polygyrus antigen extracts and provided evidence that CD4+ T cells recently producing IL-4 in vivo are impaired in their cytokine response in vitro (41). This finding might, in part, explain the lower frequencies of antigen-specific IL-4 producers detected in our in vitro assays. Second, different conditions, such as the infection time point analyzed, the source of antigen-presenting cells, in vitro cell numbers during restimulation, or antigen dose, might have led to an underestimation of specific CD4 T-cell numbers based on CD154 expression. However, despite the marked difference between the frequencies of antigen-specific CD4+ T cells presented here and for the IL-4 reporter system, our data show that the CD154 technique facilitates the analysis of a broader spectrum of pathogen-specific cytokine responses in a nonmanipulated immune system.

Adoptive transfer of CD4 cells did not show an effect on the worm burden (data not shown). This observation further supports the concept that resistance to gastrointestinal helminths is dependent on CD4+ Th2-type immune responses, as shown in animal models with H. polygyrus or Nippostrongylus brasiliensis (11, 64, 65), as well as in humans infected with Ascaris lumbricoides and Trichuris trichiura (24, 62).

Hence, our study is in line with the recent finding that memory CD4+ T cells develop after infection with the gastrointestinal nematode Trichuris muris and mediate protection against rechallenge (69). In a recent publication introducing alternatively activated macrophages as an effector population essential for protective immunity to challenge infections with H. polygyrus, it was shown that memory Th2 cells derived from H. polygyrus-cured mice transferred protection against a primary infection (4). The protective effect of Th2 memory cells was more pronounced than what we found in our transfer model using Teff cells from an ongoing infection. It would be interesting to investigate whether differences in, e.g., the homing receptor repertoires or susceptibility to induced cell death between memory and effector T cells might be responsible for this phenomenon.

In conclusion, our data indicate that in our model system, tipping the balance of effector T cells during infection strongly influences the survival of parasitic nematodes, while Treg cells may have functions that are not directly related to worm persistence.

Acknowledgments

The study was supported by Deutsche Forschungsgemeinschaft SFB 650 to S.H., R.L., J.H., and A.H.

We thank B. Sonnenburg and M. Müller for excellent technical assistance and K. Raba and T. Kaiser for FACS sorting.

Editor: J. F. Urban, Jr.

Footnotes

Published ahead of print on 3 March 2008.

REFERENCES

  • 1.Akiho, H., Y. Deng, P. Blennerhassett, H. Kanbayashi, and S. M. Collins. 2005. Mechanisms underlying the maintenance of muscle hypercontractility in a model of postinfective gut dysfunction. Gastroenterology 129131-141. [DOI] [PubMed] [Google Scholar]
  • 2.Al-Qaoud, K. M., B. Fleischer, and A. Hoerauf. 1998. The Xid defect imparts susceptibility to experimental murine filariosis—association with a lack of antibody and IL-10 production by B cells in response to phosphorylcholine. Int. Immunol. 1017-25. [DOI] [PubMed] [Google Scholar]
  • 3.Anderson, C. F., M. Oukka, V. J. Kuchroo, and D. Sacks. 2007. CD4+CD25Foxp3 Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. J. Exp. Med. 204285-297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Anthony, R. M., J. F. Urban, Jr., F. Alem, H. A. Hamed, C. T. Rozo, J. L. Boucher, N. Van Rooijen, and W. C. Gause. 2006. Memory TH2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat. Med. 12955-960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Babu, S., C. P. Blauvelt, V. Kumaraswami, and T. B. Nutman. 2006. Regulatory networks induced by live parasites impair both TH1 and TH2 pathways in patent lymphatic filariasis: implications for parasite persistence. J. Immunol. 1763248-3256. [DOI] [PubMed] [Google Scholar]
  • 6.Baumgart, M., F. Tompkins, J. Leng, and M. Hesse. 2006. Naturally occurring CD4+Foxp3+ regulatory T cells are an essential, IL-10-independent part of the immunoregulatory network in Schistosoma mansoni egg-induced inflammation. J. Immunol. 1765374-5387. [DOI] [PubMed] [Google Scholar]
  • 7.Behnke, J. M., A. Lowe, S. Clifford, and D. Wakelin. 2003. Cellular and serological responses in resistant and susceptible mice exposed to repeated infection with Heligmosomoides polygyrus bakeri. Parasite Immunol. 25333-340. [DOI] [PubMed] [Google Scholar]
  • 8.Beiting, D. P., L. F. Gagliardo, M. Hesse, S. K. Bliss, D. Meskill, and J. A. Appleton. 2007. Coordinated control of immunity to muscle stage Trichinella spiralis by IL-10, regulatory T cells, and TGF-beta. J. Immunol. 1781039-1047. [DOI] [PubMed] [Google Scholar]
  • 9.Belkaid, Y., C. A. Piccirillo, S. Mendez, E. M. Shevach, and D. L. Sacks. 2002. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature 420502-507. [DOI] [PubMed] [Google Scholar]
  • 10.Doligalska, M., K. Donskow-Schmelter, J. Rzepecka, and N. Drela. 2007. Reduced apoptosis in BALB/c mice infected with Heligmosomoides polygyrus. Parasite Immunol. 29283-291. [DOI] [PubMed] [Google Scholar]
  • 11.Fallon, P. G., H. E. Jolin, P. Smith, C. L. Emson, M. J. Townsend, R. Fallon, P. Smith, and A. N. McKenzie. 2002. IL-4 induces characteristic Th2 responses even in the combined absence of IL-5, IL-9, and IL-13. Immunity 177-17. [DOI] [PubMed] [Google Scholar]
  • 12.Finkelman, F. D., T. Shea-Donohue, J. Goldhill, C. A. Sullivan, S. C. Morris, K. B. Madden, W. C. Gause, and J. F. Urban, Jr. 1997. Cytokine regulation of host defense against parasitic gastrointestinal nematodes: lessons from studies with rodent models. Annu. Rev. Immunol. 15505-533. [DOI] [PubMed] [Google Scholar]
  • 13.Finkelman, F. D., T. Shea-Donohue, S. C. Morris, L. Gildea, R. Strait, K. B. Madden, L. Schopf, and J. F. Urban, Jr. 2004. Interleukin-4- and interleukin-13-mediated host protection against intestinal nematode parasites. Immunol. Rev. 201139-155. [DOI] [PubMed] [Google Scholar]
  • 14.Finney, C. A., M. D. Taylor, M. S. Wilson, and R. M. Maizels. 2007. Expansion and activation of CD4+CD25+ regulatory T cells in Heligmosomoides polygyrus infection. Eur. J. Immunol. 371874-1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Frentsch, M., O. Arbach, D. Kirchhoff, B. Moewes, M. Worm, M. Rothe, A. Scheffold, and A. Thiel. 2005. Direct access to CD4+ T cells specific for defined antigens according to CD154 expression. Nat. Med. 111118-1124. [DOI] [PubMed] [Google Scholar]
  • 16.Gessner, A., K. Mohrs, and M. Mohrs. 2005. Mast cells, basophils, and eosinophils acquire constitutive IL-4 and IL-13 transcripts during lineage differentiation that are sufficient for rapid cytokine production. J. Immunol. 1741063-1072. [DOI] [PubMed] [Google Scholar]
  • 17.Gillan, V., R. A. Lawrence, and E. Devaney. 2005. B cells play a regulatory role in mice infected with the L3 of Brugia pahangi. Int. Immunol. 17373-382. [DOI] [PubMed] [Google Scholar]
  • 18.Grencis, R. K., and A. J. Bancroft. 2004. Interleukin-13: a key mediator in resistance to gastrointestinal-dwelling nematode parasites. Clin. Rev. Allergy Immunol. 2651-60. [DOI] [PubMed] [Google Scholar]
  • 19.Harnett, W., and M. M. Harnett. 2006. What causes lymphocyte hyporesponsiveness during filarial nematode infection? Trends Parasitol. 22105-110. [DOI] [PubMed] [Google Scholar]
  • 20.Hesse, M., C. A. Piccirillo, Y. Belkaid, J. Prufer, M. Mentink-Kane, M. Leusink, A. W. Cheever, E. M. Shevach, and T. A. Wynn. 2004. The pathogenesis of schistosomiasis is controlled by cooperating IL-10-producing innate effector and regulatory T cells. J. Immunol. 1723157-3166. [DOI] [PubMed] [Google Scholar]
  • 21.Hisaeda, H., Y. Maekawa, D. Iwakawa, H. Okada, K. Himeno, K. Kishihara, S. Tsukumo, and K. Yasutomo. 2004. Escape of malaria parasites from host immunity requires CD4+CD25+ regulatory T cells. Nat. Med. 1029-30. [DOI] [PubMed] [Google Scholar]
  • 22.Huehn, J., and A. Hamann. 2005. Homing to suppress: address codes for Treg migration. Trends. Immunol. 26632-636. [DOI] [PubMed] [Google Scholar]
  • 23.Huehn, J., K. Siegmund, J. C. Lehmann, C. Siewert, U. Haubold, M. Feuerer, G. F. Debes, J. Lauber, O. Frey, G. K. Przybylski, U. Niesner, M. de la Rosa, C. A. Schmidt, R. Brauer, J. Buer, A. Scheffold, and A. Hamann. 2004. Developmental stage, phenotype, and migration distinguish naive- and effector/memory-like CD4+ regulatory T cells. J. Exp. Med. 199303-313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jackson, J. A., J. D. Turner, L. Rentoul, H. Faulkner, J. M. Behnke, M. Hoyle, R. K. Grencis, K. L. Else, J. Kamgno, M. Boussinesq, and J. E. Bradley. 2004. T helper cell type 2 responsiveness predicts future susceptibility to gastrointestinal nematodes in humans. J. Infect. Dis. 1901804-1811. [DOI] [PubMed] [Google Scholar]
  • 25.Jankovic, D., M. C. Kullberg, C. G. Feng, R. S. Goldszmid, C. M. Collazo, M. Wilson, T. A. Wynn, M. Kamanaka, R. A. Flavell, and 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. 204273-283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kirchhoff, D., M. Frentsch, P. Leclerk, D. Bumann, S. Rausch, S. Hartmann, A. Thiel, and A. Scheffold. 2007. Identification and isolation of murine antigen-reactive T cells according to CD154 expression. Eur. J. Immunol. 372370-2377. [DOI] [PubMed] [Google Scholar]
  • 27.Kitagaki, K., T. R. Businga, D. Racila, D. E. Elliott, J. V. Weinstock, and J. N. Kline. 2006. Intestinal helminths protect in a murine model of asthma. J. Immunol. 1771628-1635. [DOI] [PubMed] [Google Scholar]
  • 28.Kursar, M., M. Koch, H. W. Mittrucker, G. Nouailles, K. Bonhagen, T. Kamradt, and S. H. Kaufmann. 2007. Regulatory T cells prevent efficient clearance of Mycobacterium tuberculosis. J. Immunol. 1782661-2665. [DOI] [PubMed] [Google Scholar]
  • 29.Layland, L. E., R. Rad, H. Wagner, and C. U. da Costa. 2007. Immunopathology in schistosomiasis is controlled by antigen-specific regulatory T cells primed in the presence of TLR2. Eur. J. Immunol. 372174-2184. [DOI] [PubMed] [Google Scholar]
  • 30.Lehmann, J., Huehn, J., M. de la Rosa, F. Maszyna, U. Kretschmer, V. Krenn, M. Brunner, A. Scheffold, and A. Hamann. 2002. Expression of the integrin αEβ7 identifies unique subsets of CD25+ as well as CD25 regulatory T cells. Proc. Natl. Acad. Sci. USA 9913031-13036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Livak, K. J., and T. D. Schmittgen. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 25402-408. [DOI] [PubMed] [Google Scholar]
  • 32.Loddenkemper, C., J. Maul, E. Berg, H. Stein, M. Zeitz, and R. Duchmann. 2006. Analysis of FOXP3 protein expression in human CD4+CD25+ regulatory T cells at the single-cell level. Eur. J. Immunol. 36245. [DOI] [PubMed] [Google Scholar]
  • 33.Loddenkemper, C., M. Schernus, M. Noutsias, H. Stein, E. Thiel, and D. Nagorsen. 2006. In situ analysis of FOXP3+ regulatory T cells in human colorectal cancer. J. Transl. Med. 452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Maizels, R. M., and M. Yazdanbakhsh. 2003. Immune regulation by helminth parasites: cellular and molecular mechanisms. Nat. Rev. Immunol. 3733-744. [DOI] [PubMed] [Google Scholar]
  • 35.Maizels, R. M., A. Balic, N. Gomez-Escobar, M. Nair, M. D. Taylor, and J. E. Allen. 2004. Helminth parasites: masters of regulation. Immunol. Rev. 20189-116. [DOI] [PubMed] [Google Scholar]
  • 36.Maloy, K. J., L. Salaun, R. Cahill, G. Dougan, N. J. Saunders, and F. Powrie. 2003. CD4+CD25+ T(R) cells suppress innate immune pathology through cytokine-dependent mechanisms. J. Exp. Med. 197111-119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.McKee, A. S., and E. J. Pearce. 2004. CD25+CD4+ cells contribute to Th2 polarization during helminth infection by suppressing Th1 response development. J. Immunol. 1731224-1231. [DOI] [PubMed] [Google Scholar]
  • 38.Mendez, S., S. K. Reckling, C. A. Piccirillo, D. Sacks, and Y. Belkaid. 2004. Role for CD4+ CD25+ regulatory T cells in reactivation of persistent leishmaniasis and control of concomitant immunity. J. Exp. Med. 200201-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Metwali, A., T. Setiawan, A. M. Blum, J. Urban, D. E. Elliott, L. Hang, and J. V. Weinstock. 2006. Induction of CD8+ regulatory T cells in the intestine by Heligmosomoides polygyrus infection. Am. J. Physiol. Gastrointest. Liver 291253-259. [DOI] [PubMed] [Google Scholar]
  • 40.Miyara, M., and S. Sakaguchi. 2007. Natural regulatory T cells: mechanisms of suppression. Trends. Mol. Med. 13108-116. [DOI] [PubMed] [Google Scholar]
  • 41.Mohrs, K., A. E. Wakil, N. Killeen, R. M. Locksley, and M. Mohrs. 2005. A two-step process for cytokine production revealed by IL-4 dual-reporter mice. Immunity 23419-429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Monroy, F. G., and F. J. Enriquez. 1992. Heligmosomoides polygyrus: a model for chronic gastrointestinal helminthiasis. Parasitol. Today 849-54. [DOI] [PubMed] [Google Scholar]
  • 43.Morimoto, M., M. Morimoto, J. Whitmire, S. Xiao, R. M. Anthony, H. Mirakami, R. A. Star, J. F. Urban, Jr., and W. C. Gause. 2004. Peripheral CD4 T cells rapidly accumulate at the host:parasite interface during an inflammatory Th2 memory response. J. Immunol. 1722424-2430. [DOI] [PubMed] [Google Scholar]
  • 44.Nie, C. Q., N. J. Bernard, L. Schofield, and D. S. Hansen. 2007. CD4+ CD25+ regulatory T cells suppress CD4+ T-cell function and inhibit the development of Plasmodium berghei-specific TH1 responses involved in cerebral malaria pathogenesis. Infect. Immun. 752275-2282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.O'Garra, A., R. Chang, N. Go, R. Hastings, C. Haughton, and M. Howard. 1992. Ly-1 B (B-1) cells are the main source of B cell-derived interleukin 10. Eur. J. Immunol. 22711-717. [DOI] [PubMed] [Google Scholar]
  • 46.Palanivel, V., C. Posey, A. M. Horauf, W. Solbach, W. F. Piessens, and D. A. Harn. 1996. B-cell outgrowth and ligand-specific production of IL-10 correlate with Th2 dominance in certain parasitic diseases. Exp. Parasitol. 84168-177. [DOI] [PubMed] [Google Scholar]
  • 47.Pritchard, D. I., C. E. Lawrence, P. Appleby, I. A. Gibb, and K. Glover. 1994. Immunosuppressive proteins secreted by the gastrointestinal nematode parasite Heligmosomoides polygyrus. Int. J. Parasitol. 24495-500. [DOI] [PubMed] [Google Scholar]
  • 48.Raghavan, S., and J. Holmgren. 2005. CD4+CD25+ suppressor T cells regulate pathogen induced inflammation and disease. FEMS Immunol. Med. Microbiol. 44121-127. [DOI] [PubMed] [Google Scholar]
  • 49.Rzepecka, J., R. Lucius, M. Doligalska, S. Beck, S. Rausch, and S. Hartmann. 2006. Screening for immunomodulatory proteins of the intestinal parasitic nematode Heligmosomoides polygyrus. Parasite Immunol. 28463-472. [DOI] [PubMed] [Google Scholar]
  • 50.Sakaguchi, S., and F. Powrie. 2007. Emerging challenges in regulatory T cell function and biology. Science 317627-629. [DOI] [PubMed] [Google Scholar]
  • 51.Satoguina, J., M. Mempel, J. Larbi, M. Badusche, C. Loliger, O. Adjei, G. Gachelin, B. Fleischer, and A. Hoerauf. 2002. Antigen-specific T regulatory-1 cells are associated with immunosuppression in a chronic helminth infection (onchocerciasis). Microbes Infect. 41291-1300. [DOI] [PubMed] [Google Scholar]
  • 52.Segura, M., Z. Su, C. Piccirillo, and M. M. Stevenson. 2007. Impairment of dendritic cell function by excretory-secretory products: a potential mechanism for nematode-induced immunosuppression. Eur. J. Immunol. 371887-1904. [DOI] [PubMed] [Google Scholar]
  • 53.Shea-Donohue, T., C. Sullivan, F. D. Finkelman, K. B. Madden, S. C. Morris, J. Goldhill, V. Pineiro-Carrero, and J. F. Urban, Jr. 2001. The role of IL-4 in Heligmosomoides polygyrus-induced alterations in murine intestinal epithelial cell function. J. Immunol. 1672234-2239. [DOI] [PubMed] [Google Scholar]
  • 54.Siegmund, K., M. Feuerer, C. Siewert, S. Ghani, U. Haubold, A. Dankof, V. Krenn, M. P. Schon, A. Scheffold, J. B. Lowe, A. Hamann, U. Syrbe, and J. Huehn. 2005. Migration matters: regulatory T-cell compartmentalization determines suppressive activity in vivo. Blood 1063097-3104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Solbach, W., and R. Lucius. 2005. Parasite evasion, p. 675-691. In S. Kaufmann and M. Steward (ed.), Topley and Wilson microbiology and microbial infections, 10th ed. Hodder Arnold, London, United Kingdom.
  • 56.Su, Z., M. Segura, K. Morgan, J. C. Loredo-Osti, and M. M. Stevenson. 2005. Impairment of protective immunity to blood-stage malaria by concurrent nematode infection. Infect. Immun. 733531-3539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Suffia, I., S. K. Reckling, G. Salay, and Y. Belkaid. 2005. A role for CD103 in the retention of CD4+CD25+ Treg and control of Leishmania major infection. J. Immunol. 1745444-5455. [DOI] [PubMed] [Google Scholar]
  • 58.Taylor, J. D., M. Mohrs, and E. J. Pearce. 2006. Regulatory T cell responses develop in parallel to Th responses and control the magnitude and phenotype of the Th effector population. J. Immunol. 1765839-5847. [DOI] [PubMed] [Google Scholar]
  • 59.Taylor, M. D., L. LeGoff, A. Harris, E. Malone, J. E. Allen, and R. M. Maizels. 2005. Removal of regulatory T cell activity reverses hyporesponsiveness and leads to filarial parasite clearance in vivo. J. Immunol. 1744924-4933. [DOI] [PubMed] [Google Scholar]
  • 60.Taylor, M. D., A. Harris, M. G. Nair, R. M. Maizels, and J. E. Allen. 2006. F4/80+ alternatively activated macrophages control CD4+ T cell hyporesponsiveness at sites peripheral to filarial infection. J. Immunol. 1766918-6927. [DOI] [PubMed] [Google Scholar]
  • 61.Trinchieri, G. 2007. Interleukin-10 production by effector T cells: Th1 cells show self control. J. Exp. Med. 204239-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Turner, J. D., H. Faulkner, J. Kamgno, F. Cormont, J. van Snick, K. J. Else, R. K. Grencis, J. M. Behnke, M. Boussinesq, and J. E. Bradley. 2003. Th2 cytokines are associated with reduced worm burdens in a human intestinal helminth infection. J. Infect. Dis. 1881768-1775. [DOI] [PubMed] [Google Scholar]
  • 63.Urban, J. F., Jr., I. M. Katona, W. E. Paul, and F. D. Finkelman. 1991. Interleukin 4 is important in protective immunity to a gastrointestinal nematode infection in mice. Proc. Natl. Acad. Sci. USA 885513-5517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Urban, J. F., Jr., C. R. Maliszewski, K. B. Madden, I. M. Katona, and F. D. Finkelman. 1995. IL-4 treatment can cure established gastrointestinal nematode infections in immunocompetent and immunodeficient mice. J. Immunol. 1544675-4684. [PubMed] [Google Scholar]
  • 65.Urban, J. F., Jr., N. Noben-Trauth, D. D. Donaldson, K. B. Madden, S. C. Morris, M. Collins, and F. D. Finkelman. 1998. IL-13, IL-4Rα, and Stat6 are required for the expulsion of the gastrointestinal nematode parasite Nippostrongylus brasiliensis. Immunity 8255-264. [DOI] [PubMed] [Google Scholar]
  • 66.Uzonna, J. E., G. Wei, D. Yurkowski, and P. Bretscher. 2001. Immune elimination of Leishmania major in mice: implications for immune memory, vaccination, and reactivation disease. J. Immunol. 1676967-6974. [DOI] [PubMed] [Google Scholar]
  • 67.Walther, M., J. E. Tongren, L. Andrews, D. Korbel, E. King, H. Fletcher, R. F. Andersen, P. Bejon, F. Thompson, S. J. Dunachie, F. Edele, J. B. de Souza, R. E. Sinden, S. C. Gilbert, E. M. Riley, and A. V. Hill. 2005. Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity 23287-296. [DOI] [PubMed] [Google Scholar]
  • 68.Wilson, M. S., M. D. Taylor, A. Balic, C. A. Finney, J. R. Lamb, and R. M. Maizels. 2005. Suppression of allergic airway inflammation by helminth-induced regulatory T cells. J. Exp. Med. 2021199-1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zaph, C., K. A. Rook, M. Goldschmidt, M. Mohrs, P. Scott, and D. Artis. 2006. Persistence and function of central and effector memory CD4+ T cells following infection with a gastrointestinal helminth. J. Immunol. 177511-518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhao, A., J. McDermott, J. F. Urban, Jr., W. Gause, K. B. Madden, K. A. Yeung, S. C. Morris, F. D. Finkelman, and T. Shea-Donohue. 2003. Dependence of IL-4, IL-13, and nematode-induced alterations in murine small intestinal smooth muscle contractility on Stat6 and enteric nerves. J. Immunol. 171948-954. [DOI] [PubMed] [Google Scholar]

Articles from Infection and Immunity are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES