Helminth parasites provoke multicellular immune responses in their hosts that can suppress concomitant disease. The gut lumen-dwelling tapeworm Hymenolepis diminuta, unlike other parasites assessed as helminth therapy, causes no host tissue damage while potently suppressing murine colitis. With the goal of harnessing the immunomodulatory capacity of infection with H. diminuta, we assessed the putative generation of anti-colitic regulatory B cells following H. diminuta infection. Splenic CD19+ B cells isolated from mice infected 7 [HdBc(7d)] and 14 d (but not 3 d) previously with H. diminuta and transferred to naive mice significantly reduced the severity of dinitrobenzene sulfonic acid (DNBS)-, oxazolone-, and dextran-sodium sulfate–induced colitis. Mechanistic studies with the DNBS model, revealed the anti-colitic HdBc(7d) was within the follicular B cell population and its phenotype was not dependent on IL-4 or IL-10. The HdBc(7d) were not characterized by increased expression of CD1d, CD5, CD23, or IL-10 production, but did spontaneously, and upon LPS plus anti-CD40 stimulation, produce more TGF-β than CD19+ B cells from controls. DNBS-induced colitis in RAG1−/− mice was inhibited by administration of HdBc(7d), indicating a lack of a requirement for T and B cells in the recipient; however, depletion of macrophages in recipient mice abrogated the anti-colitic effect of HdBc(7d). Thus, in response to H. diminuta, a putatively unique splenic CD19+ B cell with a functional immunoregulatory program is generated that promotes the suppression of colitis dominated by TH1, TH2, or TH1-plus-TH2 events, and may do so via the synthesis of TGF-β and the generation of, or cooperation with, a regulatory macrophage.

Helminth parasites have developed efficient strategies to circumvent their host’s efforts to identify and destroy or expel them, allowing their long-term survival often without compromising host survival (1, 2). Moreover, despite the variety in helminth life cycles and preferred niches in mammalian hosts, they generally induce stereotypic immune responses in mice and humans that are dominated by T helper 2 (TH2) responses (3). Consequently, infection with nematode, trematode, or cestode parasites has been shown to reduce the severity of TH1-mediated inflammatory disease in rodent model systems (47); however, TH2-mediated allergic-type diseases can also be suppressed by infection with helminth parasites (8, 9). Thus, TH1-TH2 reciprocal antagonism does not fully explain the amelioration of a range of inflammatory diseases in murine model systems by infection with helminth parasites (10). Infection with helminth parasites can also reduce the severity of concomitant autoinflammatory disease in humans (11); therefore, better knowledge of immunoregulation in helminth-rodent models could pave the way for new approaches to conditions such as inflammatory bowel disease, diabetes, and airway inflammation.

It has become apparent that immunoregulatory cells are consistently mobilized in response to infection with helminths, including but not restricted to alternatively activated macrophages (AAMs) (12), Foxp3+ regulatory T cells (Tregs) (13, 14), and IL-10–producing B cells (15). For example, Foxp3+ Tregs and regulatory dendritic cells have been implicated in the suppression of murine colitis that accompanies infection with the intestinal nematode Heligmosomoides polygyrus (16, 17), and macrophages recruited to the lamina propria after infection with Schistosoma mansoni inhibited dextran sodium sulfate (DSS)-induced colitis (18).

Hymenolepis diminuta is a largely innocuous cestode that is expelled from mice within 12–14 d of a primary infection (19). Infection with H. diminuta significantly reduces the severity of dinitrobenzene sulfonic acid (DNBS)-induced colitis in mice, an effect that involved IL-10 likely from CD4+ T cells. Extrapolating from the observation of increased expression of markers of AAMs in the gut of parasitized mice, we showed that adoptive transfer of ex vivo–differentiated AAMs inhibited DNBS-induced colitis (20). Because the immune response to combat helminths is a coordinated multicellular one, the accompanying or subsequent immunoregulatory events are likely to be equally complex; therefore, it is highly likely that other immunoregulatory cells, in addition to CD4+ T cells and AAMs, are mobilized after infection with H. diminuta.

Subpopulations of B cells with immunosuppressive properties have been identified (21); for example, B cells that arise in response to infection with helminths can suppress airway inflammation and murine experimental autoimmune encephalitis, which is a model of multiple sclerosis (2224). There is, at best, a rudimentary knowledge of regulatory B cell subtypes that arise upon infection with helminths, noting that the few studies available have used nematodes or the trematode S. mansoni. Thus, in ongoing analyses to dissect the multiple mechanisms by which infection with helminth parasites can suppress colitis, the current study assessed the nature and potential anti-inflammatory effect of splenic B cells harvested from mice after a primary infection with H. diminuta.

Male BALB/c or C57BL/6 mice (7–8 wk old) were purchased from Charles River Laboratories (Quebec, Canada). IL-4 receptor-α knockout (KO) mice (BALB/c background) were a gift from Dr. F. Brombacher (University of Cape Town, South Africa), RAG1 KO-mice (C57BL/6 background; lack T and B cells) and mast cell deficient (kitw-sh) mice were from breeding colonies provided by Dr. P. Santamaria and Dr. M. Kelly (University of Calgary). IL-10 KO (BALB/c background) and additional RAG1−/− mice were purchased from Jackson Laboratories. Mice were housed under a light-dark cycle of 12:12 h with free access to food and water. Animal experiments were conducted with approval from the University of Calgary Health Science Animal Care Committee conforming to national guidelines.

The life cycle of H. diminuta was maintained in the laboratory by cyclical passage through the intermediate invertebrate host, the flour beetle, Tribolium confusum. Mice were infected with five cysticercoids in 100 μl of sterile 0.9% NaCl by oral gavage (25).

Splenocytes were isolated from mice infected with H. diminuta 3, 7 [HdBc(7d)] or 14 d previously, and CD19+ B cells were obtained using magnetic separation and a mouse B cell negative selection kit (StemCell Technologies, Vancouver, Canada) following the manufacturer’s instructions. Purified splenic B cells were washed in sterile PBS, and 5 × 106 B cells (90–92% CD19+) or 3 × 106 follicular B cells (see below) were given i.p. to recipient mice immediately after the intrarectal (i.r.) delivery of either DNBS or oxazolone (pilot studies with 3 × 106/ml splenic CD19+ cells produced inconsistent data) or when regular drinking water was replaced with 5% w/v DSS-drinking water. In addition, B cells from naive mice were treated in vitro for 48 h with either murine recombinant IL-4 plus IL-13 (both 10 ng/ml; Cedarlane Laboratories, Burlington, ON, Canada; referred to as BcIL-4/13) or 1 mg/ml of a PBS-soluble crude Ag extract of whole adult H. diminuta (referred to as HdAg) (26). The cells were rinsed extensively with PBS and used in transfer experiments.

Experiments were performed to assess any relevant participation of the 8–10% contaminant cells after magnetic purification. Cell sorting (FACSAria II machine and FACSDiva version 6.1.3. software, BD Biosciences; at University of Calgary flow cytometry facilities) was performed, yielding a 99.1% CD19+ cell population. Next, splenic follicular B cells were isolated by cell sorting based on CD23+IgMlo surface expression (27).

To track HdBc(7d) after transfer, cells were stained with CellTrace CFSE labeling kit (Molecular Probes). Splenocytes were adjusted to 5 × 106/ml PBS/BSA 0.01% and stained with CellTrace CFSE for 10 min at 37°C. The reaction was quenched with FBS-medium, cells were spun down, and B cells were purified as above.

In other experiments, mice treated with DNBS ± HdBc(7d) also received anti–TGF-β neutralizing Abs (200 μg. i.v.; R&D Systems) immediately after i.r. delivery of DNBS and colitis assessed 72 h after DNBS administration.

Colitis was induced in anesthetized mice by i.r. DNBS (3 mg) or oxazolone (5 mg; both from Sigma, St. Louis, MO) in 100 μl of 50% ethanol via a 3-cm catheter or by a 5-d treatment with 5% DSS (MP Biochemicals, Solon, OH) (25, 28). Mice were sacrificed at 72 h after DNBS–oxazolone administration, and disease severity was assessed with a five-point disease activity score, based on animal weight and general condition, colon length, diarrhea, and macroscopic evidence of ulceration. At necropsy, the colon was removed, measured, and divided based on percentage length, such that: 1) the terminal 20% was snap-frozen for the determination of myeloperoxidase (MPO) activity using a colorimetric assay in which 1 U of MPO enzyme activity is required to degrade 1 μM H2O2 per minute; 2) the next 10% was fixed in 10% neutral buffered formalin, and histopathology was assessed after processing using a 12-point scale on H&E-stained sections in a blinded fashion; and 3) the next 10% was snap frozen in liquid nitrogen for either extraction of mRNA for quantitative polymerase chain reaction (qPCR) analysis or homogenized in PBS containing a protease inhibitor mixture (Roche, Mississauga, ON, Canada) and cytokine levels measured by ELISA (28).

Monoarthritis was induced via the intra-articular injection of 10 μl CFA (Sigma) into the ipsilateral joint and 10 μl sterile PBS into the contralateral joint as control. Knee joint swelling was monitored over a 6d period by an observer blinded to the experimental treatment using mechanical Vernier calipers as described previously (6).

Total cells were gated in a forward light scatter–versus-CD19 plot to identify B cells, and specific surface markers were analyzed. The cell surface Ags CD5 and CD1d are prominent markers of regulatory B cells, and Tim-1 (29) and CD23 (24) have also been associated with specific regulatory B cell subtypes. One million B cells were treated with anti-CD16/32 (10 min, 4°C) to block nonspecific binding, and then standard flow cytometry staining protocols were conducted with the following fluorochrome-conjugated Abs: APC-CD19, FITC-CD5, PE-CD1d, PE-Tim1, PE-CD23, PE-CD138, PE-CD80, PerCP-MHC II, FITC-B220, FITC-IgM, PE-programmed death ligand (PDL)-1, and PE-PDL-2 (all from BioLegend, San Diego, CA). APC-F4/80 and PE-Gr1 were used as markers of macrophages and neutrophils, respectively. Data were analyzed using the Attune flow cytometer and Attune cytometric software version 1.2.5 (R&D systems).

Colonic tissue was homogenized in TRIzol reagent (Life Technologies, Carlsbad, CA). Total RNA was quantified using a NanoDrop (Thermo Fischer Scientific), and 1 μg isolated RNA was used to generate cDNA with an iScript RT Kit (Bio-Rad, Mississauga, ON, Canada) in a MyCycler thermocycler (Bio-Rad). cDNA was added to 500 nM gene-specific primers for murine chitinase 3–like protein (Ym1), resistin-like molecule α (Relmα), choline acetyltransferase (ChAT), and Foxp3, IL-1β, IFNγ, IL-17, and IL-10 to a 1× SYBR green reaction mix (Bio-Rad). Data were analyzed using the 2−ΔCt method using 18S as a housekeeping gene.

Isolated splenocytes (1 × 106/ml) were stimulated with Con A (2 μg/ml) for 48 h, and supernatants were collected. A panel of cytokines were screened in the Luminex 32-plex (Eve Technologies, University of Calgary). Supernatants from CD19+ B cells (1 × 106) stimulated in vitro with LPS (1 μg/ml) ± anti-CD40 (1 μg/ml) for 48 h were assessed for IgG1, IL-4, IL-10, and TGF-β by sandwich ELISA (R&D Systems) and NO by the Griess reaction (measures nitrite as a surrogate for NO). Macrophage (MØ) production of TNF-α was assessed with ELISA.

Bone marrow MØ were grown as described (30). Femur and tibias from naive BALB/c mice were flushed with sterile RPMI medium containing HEPES (20 mM), penicillin-streptomycin (2%), and 1× Glutamax (all from Life Technologies). Total bone marrow cells were treated with M-CSF (20 ng/ml; Cedarlane Laboratories, Burlington, ON) on days 0, +2 and +4; cells were harvested at day 7, and 2.5 × 105 cells were seeded into 12-well plates. HdBc(7d) were retrieved and cultured with MØ or LPS- (100 ng/ml, 48 h) activated MØ (classically activated macrophages [CAM]) for 48h, the B cells were retrieved and assessed by flow cytometry, and spontaneous IL-10 and TGF-β production was measured with ELISA. With respect to the MØ: 1) mRNA was extracted for qPCR, 2) cells stimulated with LPS (1 μg/ml) for cytokine production, and 3) in additional studies MØ were stimulated with LPS for 24 h ± HdBc(7d) at 1:2, 1:4, and 1:8 ratios(MØ:B cells), supernatants were collected, and TNFα, IL-10, and NO were measured.

Macrophages were depleted in RAG1−/− mice using clodronate (dichloromethylene diphosphate)-containing liposomes (Roche Diagnostics, Mannheim, Germany); 200 μl of PBS or clodronate-loaded liposomes were injected i.p. 1 d before, the same day, and 1 d after DNBS ± HdBc(7d) administration (20). Efficiency of MØ depletion was assessed by detection of F4/80+ cells in the spleen and peritoneal lavage (Gr1+ used to identify neutrophils). Splenic CD19+ B cells were treated with clodronate-loaded liposomes in vitro, and viability was assessed.

Splenic CD19+ B cells from 7-d H. diminuta–infected and uninfected mice (as controls) were isolated by cell sorting (purity = 99.1%). Cells were maintained in RLT buffer (Qiagen, Valencia, CA) and RNA extraction and microarray analysis (Affymetrix GeneChip Mouse Genome Array 430.2) were conducted at Linköping University. Data were analyzed with Partek Genomics Suite 6.6 software (Partek, Chesterfield, MO) at the University of Calgary.

Data are mean ± the SEM. Multiple group comparisons were performed using one-way ANOVA followed by post hoc Tukey test, whereas comparison uninfected and H. diminuta-infected used Student t test and GraphPad Prism 5 software (La Jolla, CA). In both analyses, p < 0.05 was accepted as a statistically significant difference.

Infection with H. diminuta results in a TH2-dominated immune response (19), and enumeration revealed an ∼25% increase in CD19+/CD45R(B220)+ splenic B cells 7 d postinfection: 48.8 ± 2.8 versus 38.5 ± 1.6 × 106 (n = 5; p < 0.01). Splenic B cell purified with magnetic beads yielded a population composed of 90–92% CD19+ cells, with the remainder being predominantly CD3+ cells. This population was used in the majority of the experiments; however, it was possible that the contaminating 8–10% of CD3+ cells mediated the effect and not the B cells. Thus, cell sorting was used to isolate an ∼99.1% CD19+ splenic B cells for adoptive transfer experiments in direct comparison with the B cell population isolated by magnetic bead separation (Fig. 1).

FIGURE 1.

(AD) B cells isolated by magnetic beads or cell sorting suppress DNBS-induced colitis. Splenic B cells (5 × 106) isolated 7 d after H. diminuta infection (HdBc) by magnetic bead (mag. isol) selection (90–92% CD19+) or cell sorting (99.1% CD19+; 3 × 106) have similar capacities to suppress DNBS (3 mg, i.r., 72 h)-induced colitis in BALB/c mice (mean ± SEM; n = 4–5). (E) Original magnification ×200. *p < 0.05, #p < 0.05 compared with control (con) and DNBS only groups, respectively. L, lumen; M, muscle.

FIGURE 1.

(AD) B cells isolated by magnetic beads or cell sorting suppress DNBS-induced colitis. Splenic B cells (5 × 106) isolated 7 d after H. diminuta infection (HdBc) by magnetic bead (mag. isol) selection (90–92% CD19+) or cell sorting (99.1% CD19+; 3 × 106) have similar capacities to suppress DNBS (3 mg, i.r., 72 h)-induced colitis in BALB/c mice (mean ± SEM; n = 4–5). (E) Original magnification ×200. *p < 0.05, #p < 0.05 compared with control (con) and DNBS only groups, respectively. L, lumen; M, muscle.

Close modal

Mice treated with DNBS (i.r.) developed colitis as assessed by wasting, shortening of the colon, colonic MPO activity, disease activity score, and histopathology. Concomitant administration of splenic CD19+ B cell isolated from H. diminuta–infected mice by cell sorting or magnetic beads significantly inhibited DNBS-induced colitis (Fig. 1). The similar anti-colitic outcomes of transferring cell-sorted and magnetic bead–isolated CD19+ B cells indicated that the contaminating CD19 cells (∼4 × 105 cells) in the latter were unlikely to be driving the anti-colitic effect; therefore, subsequent experiments used CD19+ cells isolated with the magnetic bead method.

In contrast to HdBc(7d), B cells from naive mice or mice infected 3 d previously [HdBc(3d)] did not affect the severity of DNBS-induced colitis (Fig. 2). Splenic CD19+ B cells from mice infected 14 d previously with H. diminuta (n = 5) protected recipients from DNBS-induced colitis (data not shown). Adoptive transfer of HdBc(7d) significantly inhibited oxazolone- (see below) and DSS-induced colitis (n = 7–9; Supplemental Fig. 1). In accordance with the reduced MPO activity in the colon of HdBc(7d) DNBS-treated mice (Fig. 2D), these animals also had fewer Gr1+ peritoneal granulocytes (data not shown). In accordance with these data, qPCR of colonic tissue from DNBS-treated mice revealed increased IL-1β, IFN-γ, and IL-17 mRNA that was reduced in mice receiving DNBS plus HdBc(7d); there was no statistical increase in IL-10 mRNA in the latter group (Fig. 3). Con A stimulation revealed reduced IFN-γ and increased IL-4 and IL-10 production by spleen cells isolated from HdBc(7d)-treated mice compared with those from DNBS ± naive B cell–treated mice (Fig. 4A–C). In contrast, colonic levels of IL-10 were reduced in DNBS-treated mice compared with controls (Fig. 4D), independent of B cell treatment.

FIGURE 2.

Splenic B cells from H. diminuta–infected mice reduce DNBS-induced colitis. Splenic CD19+ B cells (5 × 106, i.p.) isolated from H. diminuta–infected mice 7 [HdBc(7d)] but not 3 d previously inhibit DNBS-induced (3 mg, i.r., 72 h) colitis as gauged by (A) weight loss, (B) colon length, (C) disease activity score, (D) colonic MPO activity, and (E and F) histopathology (mean ± SEM; n = 11–15 mice from four experiments). Original magnification ×200. *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively. Bc, B cells from naive mice; L, lumen, M, external muscle layers.

FIGURE 2.

Splenic B cells from H. diminuta–infected mice reduce DNBS-induced colitis. Splenic CD19+ B cells (5 × 106, i.p.) isolated from H. diminuta–infected mice 7 [HdBc(7d)] but not 3 d previously inhibit DNBS-induced (3 mg, i.r., 72 h) colitis as gauged by (A) weight loss, (B) colon length, (C) disease activity score, (D) colonic MPO activity, and (E and F) histopathology (mean ± SEM; n = 11–15 mice from four experiments). Original magnification ×200. *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively. Bc, B cells from naive mice; L, lumen, M, external muscle layers.

Close modal
FIGURE 3.

Increased local mRNA for proinflammatory cytokines is not apparent in mice treated with DNBS plus HdBc(7d). qPCR performed on mRNA from colonic extracts at or adjacent to the DNBS (3 mg, i.r., 72 h) instillation site reveals increased IL-1β, IFN-γ, and IL-17 relative to control. There were no statistically significant differences in the expression of IL-10 mRNA (mean ± SEM; n = 4–5 mice). *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively.

FIGURE 3.

Increased local mRNA for proinflammatory cytokines is not apparent in mice treated with DNBS plus HdBc(7d). qPCR performed on mRNA from colonic extracts at or adjacent to the DNBS (3 mg, i.r., 72 h) instillation site reveals increased IL-1β, IFN-γ, and IL-17 relative to control. There were no statistically significant differences in the expression of IL-10 mRNA (mean ± SEM; n = 4–5 mice). *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively.

Close modal
FIGURE 4.

Hymenolepis-induced B cells affect cytokine output in spleen cells from the recipient mice. Three days after i.r. DNBS (3 mg) and i.p. delivery of 5 × 106 CD19+ splenic B cells from naive mice (Bc) or mice infected 7 d previously with H. diminuta [HdBc(7d)], splenocytes were isolated and stimulated with concanavalin-A (2 μg/ml), and (A) IFN-γ, (B) IL-4, and (C) IL-10 were assessed with ELISA. (D) Examination of extracts of colon revealed negligible levels of IL-10 in all mice that received DNBS. (E) Splenic CD19+HdBc stimulated with LPS and anti-CD40 Abs (both 1 μg/ml) showed no enhancement of IL-10 production but (F) did display increased spontaneous production of TGF-β compared with control that was not substantially increased by LPS plus anti-CD40 challenge (mean ± SEM; n = 5–6 mice). *p < 0.05, #p < 0.05 compared with control (con; or indicated group) and DNBS, respectively.

FIGURE 4.

Hymenolepis-induced B cells affect cytokine output in spleen cells from the recipient mice. Three days after i.r. DNBS (3 mg) and i.p. delivery of 5 × 106 CD19+ splenic B cells from naive mice (Bc) or mice infected 7 d previously with H. diminuta [HdBc(7d)], splenocytes were isolated and stimulated with concanavalin-A (2 μg/ml), and (A) IFN-γ, (B) IL-4, and (C) IL-10 were assessed with ELISA. (D) Examination of extracts of colon revealed negligible levels of IL-10 in all mice that received DNBS. (E) Splenic CD19+HdBc stimulated with LPS and anti-CD40 Abs (both 1 μg/ml) showed no enhancement of IL-10 production but (F) did display increased spontaneous production of TGF-β compared with control that was not substantially increased by LPS plus anti-CD40 challenge (mean ± SEM; n = 5–6 mice). *p < 0.05, #p < 0.05 compared with control (con; or indicated group) and DNBS, respectively.

Close modal

In vitro stimulation of the CD19+ B cells with LPS revealed few differences in cytokine output between the groups (Supplemental Fig. 2). Anti-CD40 plus LPS stimulation did not reveal enhanced IL-10 production by HdBc(7d) (Fig. 4E), but there was an ∼3-fold statistically significant spontaneous release of TGF-β by these cells; this was not further enhanced by anti-CD40 plus LPS (Fig. 4F). Cotreatment with anti–TGF-β Abs significantly reduced the anti-colitic effect of HdBc(7d); however, mice treated with only DNBS had enhanced disease when given TGF-β–neutralizing Abs (Fig. 5).

FIGURE 5.

Treatment with anti–TGF-β neutralizing Abs blocks the anti-colitic effect of HdBc(7d) but also enhances DNBS-induced colitis. (A) Disease activity scores demonstrating that mice treated with anti–TGF-β Ab (αTGFβ; 200 μg/mouse) show increase disease severity of DNBS-induced colitis (3 mg, i.r., 72 h). (B) Representative images of the colon (mean ± SEM; n = 5 mice). *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively.

FIGURE 5.

Treatment with anti–TGF-β neutralizing Abs blocks the anti-colitic effect of HdBc(7d) but also enhances DNBS-induced colitis. (A) Disease activity scores demonstrating that mice treated with anti–TGF-β Ab (αTGFβ; 200 μg/mouse) show increase disease severity of DNBS-induced colitis (3 mg, i.r., 72 h). (B) Representative images of the colon (mean ± SEM; n = 5 mice). *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively.

Close modal

NO has been reported to be produced by IgA+ B cells (31); however, no differences in spontaneous or stimulated NO production between HdBc(7d) and naive B cells were observed (data not shown). qPCR (n = 3–5) revealed no significant expression of ChAT mRNA in CD19+ B cells from infected or control mice. Similarly, measurement of IgG1 production after LPS plus anti-CD40 stimulation revealed no differences between the groups: naive B cell = 3.4 ± 1.5 ng/ml versus HdBc(7d) = 2.3 ± 1.3 ng/ml.

Despite the consistent and robust ability of HdBc(7d) to inhibit DNBS-induced colitis, flow cytometric analysis did not reveal any significant upregulation of CD1d, CD5, CD21, CD23, PDL1, PDL2, or Tim1 expression (markers associated with regulatory B cells [Bregs]) (21, 29) on freshly isolated cells compared with CD19+ splenic B cells from naive mice (Supplemental Fig. 3A). However, based on the total increase in splenic CD19+ B cells in infected mice this translated into a significant increase in CD19+/CD5+ cells: 6.0 ± 0.6 versus 10.2 ± 0.7 × 106 (n = 5; p = 0.002). Furthermore, adoptive transfer of 3 × 106 CD19+ follicular B cells from mice infected 7 d previously with H. diminuta resulted in significant inhibition of DNBS-induced colitis (Fig. 6).

FIGURE 6.

Splenic follicular B cells from H. diminuta–infected mice reduce DNBS-induced colitis. (A) Dot plot showing the characterization and cell sorting of splenic CD19+ follicular B cells (CD23+IgMlo) from mice infected with H. diminuta 7 d previously [FOZ HdBc(7d)]. Adoptive transfer of the FOZ HdBc (3 × 106, i.p.) concomitant with DNBS (3 mg, i.r., 72 h) suppressed colitis as gauged by (B) disease activity score, (C) colon length, (D) colonic myeloperoxidase activity, and (E and F) histopathology (mean ± SEM; n = 3–4 mice). Original magnification ×200. *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively. L, lumen, M, external muscle layers.

FIGURE 6.

Splenic follicular B cells from H. diminuta–infected mice reduce DNBS-induced colitis. (A) Dot plot showing the characterization and cell sorting of splenic CD19+ follicular B cells (CD23+IgMlo) from mice infected with H. diminuta 7 d previously [FOZ HdBc(7d)]. Adoptive transfer of the FOZ HdBc (3 × 106, i.p.) concomitant with DNBS (3 mg, i.r., 72 h) suppressed colitis as gauged by (B) disease activity score, (C) colon length, (D) colonic myeloperoxidase activity, and (E and F) histopathology (mean ± SEM; n = 3–4 mice). Original magnification ×200. *p < 0.05, #p < 0.05 compared with control (con) and DNBS only, respectively. L, lumen, M, external muscle layers.

Close modal

In addition, microarray-based global gene expression analysis revealed upregulation of 700 and 71 genes in HdBc(7d) compared with naive B cells when 1.5 fold-changes and 1.8 fold-changes were set as the threshold for analysis, respectively (Supplemental Fig. 3B: data deposited at http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE61460). For example, calcyclin, complement component 1 q subcomponent β-polypeptide, mitochondrial ribosomal protein L52, C-type lectin domain family 2, signal transducer and activator of transcription 4, and peroxisome proliferative activated receptor γ coactivator 1α genes were significantly upregulated. Although such analysis gives no indication of protein expression or function, it attests to the activation of the HdBc(7d), and the appropriate cluster analysis could reveal a set of mRNA changes as signature molecules for the H. diminuta–induced Bregs.

The small percentage (∼2%) of CD19+ cells expressing CD138 indicated that the population was not skewed toward a plasma cell phenotype (32). Neither CD80 nor MHC II were significantly increased in freshly isolated HdBc(7d) (data not shown).

Four, 24, 48, and 72 h after i.p. injection of CFSE-labeled HdBc(7d), peritoneal lavages were obtained, the omentum was observed microscopically, and CD19+ B cells were isolated from the spleen, mesenteric lymph node (MLN), and colon. At all time-points, considerable numbers of HdBc(7d) were found in the peritoneal cavity. For example, at 72 h after treatment, 13–22% of CFSE-labeled HdBc(7d) (∼750,000 cells) remained in the peritoneal cavity (n = 3). Accumulation in the omentum was inconsistent, and appreciable numbers of labeled HdBc(7d) were below the level of detection in the spleen, MLN, and colon at this time point.

Postulating that the H. diminuta–evoked Bregs could be elicited by a direct effect of worm Ag or exposure to TH2 cytokines, CD19+ B cells from naive mice were treated in vitro with worm Ag or IL-4 plus IL-13 for 48 h and their anti-colitic potential compared with HdBc(7d). Worm Ag–treated B cells did not inhibit DNBS-induced colitis, and whereas BcIL-4/13 significantly protected against this colitis, they were, on average, less effective than HdBc(7d) (Fig. 7A, 7B). When oxazolone was used to evoke colitis, there was a statistically significant difference in the protection afforded by HdBc(7d) compared with BcIL4/13 (Fig. 7C, 7D).

FIGURE 7.

Naive B cells treated in vitro with TH2 cytokines but not H. diminuta Ag suppress colitis. Colitis was elicited by i.r. (A and B) DNBS (3 mg) or (C and D) oxazolone (oxazo.; 4 mg), and mice were treated simultaneously with B cells from mice infected with H. diminuta 7 d previously [HdBc(7d)] or B cells from naive mice treated for 48 h in vitro with either a crude PBS-soluble extract of H. diminuta (Bc-Ag; 1 mg/ml) or IL-4 plus IL-13 (both 10 ng/ml; Bc [IL4/13]). (E and F) HdBc(7d) from mast cell deficient mice (Kitw-sh) or lacking the IL-4R α-chain are as efficient as cells from WT mice at suppressing DNBS-induced colitis (mean ± SEM; n = 8–10 mice from two experiments). *p < 0.05, #p< 0.05, §p < 0.05 compared with control, DNBS or oxazo., and oxazo. plus Bc IL4/13, respectively).

FIGURE 7.

Naive B cells treated in vitro with TH2 cytokines but not H. diminuta Ag suppress colitis. Colitis was elicited by i.r. (A and B) DNBS (3 mg) or (C and D) oxazolone (oxazo.; 4 mg), and mice were treated simultaneously with B cells from mice infected with H. diminuta 7 d previously [HdBc(7d)] or B cells from naive mice treated for 48 h in vitro with either a crude PBS-soluble extract of H. diminuta (Bc-Ag; 1 mg/ml) or IL-4 plus IL-13 (both 10 ng/ml; Bc [IL4/13]). (E and F) HdBc(7d) from mast cell deficient mice (Kitw-sh) or lacking the IL-4R α-chain are as efficient as cells from WT mice at suppressing DNBS-induced colitis (mean ± SEM; n = 8–10 mice from two experiments). *p < 0.05, #p< 0.05, §p < 0.05 compared with control, DNBS or oxazo., and oxazo. plus Bc IL4/13, respectively).

Close modal

Mast cells are increased by helminth and are an important arm of the TH2 response (33); it was recently shown that they can regulate immune functions performed by myeloid derived suppressor cells (34). We tested the ability of HdBc(7d) obtained from Kitw-sh mast cell–deficient mice to regulate colitis. Splenic B cells from infected Kitw-sh mice suppressed DNBS-induced colitis to a degree comparable to cells from wild-type (WT) mice (Fig. 7E). The reciprocal direction of communication was also tested, and it was found that HdBc(7d) from WT mice when transferred into mast cell–deficient mice protected the recipients from colitis, indicating that HdBc(7d)–mast cell communication is not required for the suppression of DNBS-induced colitis (Fig. 8)

FIGURE 8.

Mast cell–deficient mice (Kitw-sh) given HdBc(7d) from WT mice are protected from DNBS-induced colitis. Analysis of (A) weight loss, (B) disease activity scores, and (C) colonic MPO activity reveal significantly less severe DNBS-induced (3 mg, i.r., 72 h) colitis in mice cotreated with HdBc(7d) (5 × 106, i.p.; mean ± SEM; n = 3–15 mice. *p < 0.05, #p < 0.05 compared with control and DNBS only, respectively.

FIGURE 8.

Mast cell–deficient mice (Kitw-sh) given HdBc(7d) from WT mice are protected from DNBS-induced colitis. Analysis of (A) weight loss, (B) disease activity scores, and (C) colonic MPO activity reveal significantly less severe DNBS-induced (3 mg, i.r., 72 h) colitis in mice cotreated with HdBc(7d) (5 × 106, i.p.; mean ± SEM; n = 3–15 mice. *p < 0.05, #p < 0.05 compared with control and DNBS only, respectively.

Close modal

Moreover, CD19+ B cells from H. diminuta–infected IL-4Rα−/− mice also inhibited DNBS-induced colitis (Fig. 7). Spleen cells from IL-4Rα−/− mice produced IL-10 (∼1 ng/ml), and although less than that from infected WT mice, this is still a substantial amount of cytokine. Thus, although IL-4 likely plays a role in educating B cells in H. diminuta–infected immunocompetent mice, signaling through the IL-4Rα is not an absolute requirement for the generation of H. diminuta–induced Bregs with the capacity to inhibit colitis. These data are consistent with the finding that IL-4Rα−/− mice infected with H. diminuta (these mice fail to expel the worm; data not shown) are somewhat protected from DNBS-induced colitis (Supplemental Fig. 4).

In contrast to these analyses of colitis, HdBc(7d) did not protect mice from the monoarthritis induced by direct intra-articular injection of CFA (data not shown).

A variety of Tregs can block colitis in murine model systems (3537), and Bregs can induce Tregs (38, 39); however, at necropsy, Foxp3 mRNA expression was not increased in colonic tissue, spleen, or MLNs of mice treated with DNBS plus HdBc(7d) (Fig. 9A). To test a role for other Tregs or involvement of the recipient’s T or B cells in the suppression of disease by HdBc(7d), colitis was elicited in RAG1−/− mice. Reconstitution of RAG1−/− mice with naive B cells did not affect the outcome of DNBS-induced colitis, whereas mice given HdBc(7d) developed significantly less macroscopic and histologic disease (Fig. 9B, 9C), indicating that T and B cells in recipient mice are dispensable in the anti-colitic effect driven by HdBc(7d). Flow cytometry analysis of spleen cells from the RAG1−/− recipients failed to detect any transferred CFSE-labeled CD19+HdBc(7d) (n = 3; data not shown).

FIGURE 9.

Adaptive immune cells in recipient mice are not required by HdBc(7d) to attenuate DNBS-induced colitis. (A) qPCR revealed no differences in the expression of the Treg marker, Foxp3, in extracts of colon, MLNs, or spleen between any of the mice (n = 9). (B and C) DNBS-induced (5 mg, i.r., 72 h) colitis in RAG1−/− mice was significantly inhibited by cotreatment with CD19+ splenic B cells from 7-d H. diminuta–infected mice [HdBc(7d)], but not by B cells from naive mice (Bc). (D and E) Transfer of HdBc(7d) from IL-10−/− mice offered similar anti-colitic protection compared with HdBc(7d) from WT mice (mean ± SEM; n = 5–10 mice from 2–3 experiments). *p < 0.05, #p < 0.05 compared with control (con) and DNBS, respectively. MPO, myeloperoxidase.

FIGURE 9.

Adaptive immune cells in recipient mice are not required by HdBc(7d) to attenuate DNBS-induced colitis. (A) qPCR revealed no differences in the expression of the Treg marker, Foxp3, in extracts of colon, MLNs, or spleen between any of the mice (n = 9). (B and C) DNBS-induced (5 mg, i.r., 72 h) colitis in RAG1−/− mice was significantly inhibited by cotreatment with CD19+ splenic B cells from 7-d H. diminuta–infected mice [HdBc(7d)], but not by B cells from naive mice (Bc). (D and E) Transfer of HdBc(7d) from IL-10−/− mice offered similar anti-colitic protection compared with HdBc(7d) from WT mice (mean ± SEM; n = 5–10 mice from 2–3 experiments). *p < 0.05, #p < 0.05 compared with control (con) and DNBS, respectively. MPO, myeloperoxidase.

Close modal

Despite the fact that the HdBc(7d) were not producing enhanced amounts of IL-10, this cytokine is implicated in helminth modulation of disease and the suppression of colitis (25, 40, 41) in immunocompetent mice. Therefore, it was important to test a role for IL-10 in the current paradigm. IL-10−/− mice expel H. diminuta (D. McKay, personal communication) and Con A stimulation of B cell–depleted splenocytes from infected IL-10−/− mice resulted in ∼2-fold and ∼9-fold increases in IL-4 and IL-5 production compared with cells from uninfected mice, respectively. Adoptive transfer of HdBc(7d) from IL-10−/− mice into BALB/c mice suppressed DNBS-induced colitis to a degree not significantly different from HdBc(7d) from WT donors, indicating that IL-10 is dispensable in the HdBc(7d) that drives the suppression of colitis (Fig. 9D, 9E).

Macrophages are a diverse population of cells that can be skewed in favor of a proinflammatory or anti-inflammatory phenotype (12, 20). Testing the hypothesis that HdBc(7d) suppression of colitis required or promoted the presence of an anti-inflammatory or proresolution MØ, experiments were conducted in mice depleted of MØ using clodronate-liposomes (MØ depletion confirmed by reduction in F4/80+ cells to <2% of the cell content of the spleen and peritoneal cavity from ∼25% and ∼22% in controls, respectively; Gr1+ cell numbers were negligibly affected by the procedure; in vitro observation indicated a lack of cytotoxic effects of clodronate liposomes on CD19+ B cells) (J.L. Reyes, personal communication).

Colitis was not appreciably different among RAG1−/− mice treated with DNBS, DNBS plus clodronate, and DNBS plus HdBc(7d) plus clodronate (Fig. 10A), whereas mice administered HdBc(7d) and the control, PBS-containing liposomes had less severe disease (Fig. 10A–C). Quantitative PCR of colon extracts for arginase-1 (not shown), Ym1, and Relmα (Fig. 10D) revealed no consistent changes in the mRNA levels of these markers for AAMs (12) between the groups, suggesting that the MØ population required for the HdBc(7d) suppression of colitis is not the IL-4/IL-13 driven AAM.

FIGURE 10.

Depletion of macrophages in recipient mice prevents the suppression of DNBS-induced colitis by B cells from H. diminuta–infected mice. RAG1−/− mice were treated with clodronate-containing liposomes (clodL) to deplete macrophages or PBS-liposomes (PBSL) as a control prior to the induction of colitis with DNBS (3 mg, i.r., 72 h) ± CD19+ B cells (5 × 106, ip.) from H. diminuta–infected mice [7 d postinfection; HdBc(7d)]. (A and B) Disease activity and histopathology scores were not different between DNBS, DNBS + clodL and DNBS + HdBc(7d)+clodL treatment groups (n = 4–9 mice from two experiments. (C) Representative histologic images. Original magnification ×200. (D) qPCR of tissue from WT mice revealed no differences in colonic mRNA expression of Ym1 or Relmα, markers indicative of alternatively activated macrophages, between the groups (n = 4). (E) Coculture of bone marrow-derived MØ with HdBc(7d) from WT mice enhances IL-10 production by LPS-stimulated MØ (100 ng/ml, 24 h; mean ± SEM; n = 6–10). *p < 0.05, #p < 0.05 compared with control (con) and DNBS group, respectively. L, gut lumen; M, external muscle layers.

FIGURE 10.

Depletion of macrophages in recipient mice prevents the suppression of DNBS-induced colitis by B cells from H. diminuta–infected mice. RAG1−/− mice were treated with clodronate-containing liposomes (clodL) to deplete macrophages or PBS-liposomes (PBSL) as a control prior to the induction of colitis with DNBS (3 mg, i.r., 72 h) ± CD19+ B cells (5 × 106, ip.) from H. diminuta–infected mice [7 d postinfection; HdBc(7d)]. (A and B) Disease activity and histopathology scores were not different between DNBS, DNBS + clodL and DNBS + HdBc(7d)+clodL treatment groups (n = 4–9 mice from two experiments. (C) Representative histologic images. Original magnification ×200. (D) qPCR of tissue from WT mice revealed no differences in colonic mRNA expression of Ym1 or Relmα, markers indicative of alternatively activated macrophages, between the groups (n = 4). (E) Coculture of bone marrow-derived MØ with HdBc(7d) from WT mice enhances IL-10 production by LPS-stimulated MØ (100 ng/ml, 24 h; mean ± SEM; n = 6–10). *p < 0.05, #p < 0.05 compared with control (con) and DNBS group, respectively. L, gut lumen; M, external muscle layers.

Close modal

In addition, in vitro coculture of HdBc(7d) with MØ for 24 or 48 h affected neither the MØ-constitutive or LPS-stimulated expression of arginase-1 and iNOS mRNA nor production of NO and TNF-α (n = 4–6; data not shown); however, IL-10 output was increased (Fig. 10E). Although IL-10 is not a requirement in the HdBc(7d), it might be an active anti-inflammatory component in the MØ effector cell.

Having identified the need for MØ in the suppression of colitis initiated by HdBc(7d), we queried the ability of MØ to affect the B cells. Coculture of HdBc(7d) with MØ resulted in significant upregulation of the B cell activation markers CD5, CD1d (that would allow interaction with NKT cells), and PDL1 (that would promote cell killing/anergy; Fig. 11), whereas levels of CD80 (Fig. 11) and CD23 (not shown) were unchanged. Coculture with CAM to mimic the likelihood of encountering LPS-exposed MØ in vivo resulted in similar changes (of lesser magnitude) in these makers on the B cells. Coculture with MØ or CAM did not affect the spontaneous production of IL-10 or TGF-β by the HdBc(7d) (n = 4; data not shown).

FIGURE 11.

HdBc(7d) are activated by coculture with MØ. Freshly isolated HdBc(7d) display no obvious upregulation of activation or Breg markers (see Supplemental Fig. 3), but after 48 h of coculture with bone-marrow derived MØ (250,000) they have increased expression of CD5 (A), CD1d, and programmed death ligand-1 (PDL1), whereas CD80 levels remained unchanged (B). A similar pattern was noted after coculture with LPS-treated MØ (1 μg/ml; mean ± SEM; n = 4. *p < 0.05 compared with medium controls.

FIGURE 11.

HdBc(7d) are activated by coculture with MØ. Freshly isolated HdBc(7d) display no obvious upregulation of activation or Breg markers (see Supplemental Fig. 3), but after 48 h of coculture with bone-marrow derived MØ (250,000) they have increased expression of CD5 (A), CD1d, and programmed death ligand-1 (PDL1), whereas CD80 levels remained unchanged (B). A similar pattern was noted after coculture with LPS-treated MØ (1 μg/ml; mean ± SEM; n = 4. *p < 0.05 compared with medium controls.

Close modal

Although B cells participate in autoimmunity, their absence can render mice susceptible to some inflammatory diseases (42). Infection with helminth parasites is a potent immunoregulatory force that can suppress inflammatory disease via many mechanisms, including mobilization of heterogeneous Breg populations that can be equally diverse in their mechanism of immune suppression. For example, suppression of inflammation by infection with S. mansoni (trematode) or H. polygyrus (nematode) is IL-10 dependent and IL-10independent, respectively (15, 24).

Expulsion of H. diminuta from mice has the bystander benefit of limiting the severity of DNBS-induced colitis (25). Given that worm expulsion is a multicellular event, many immune cell phenotypes could contribute to the suppression of colitis, and we focused on Bregs because of the paucity of data on the helminth-evoked Breg and the complete lack of data on Bregs after infection with cestodes. Splenic CD19+ B cells were increased in infected animals and adoptive transfer of HdBc(7d) and HdBc(14d), but not HdBc(3d), reduced the severity of DNBS-, oxazolone-, and DSS-induced colitis. Consistent with this, HdBc(7d) transfer resulted in reduced IL-1β and IFN-γ mRNA in the colon, and reduced IFN-γ and enhanced IL-4 and IL-10 output by splenocytes from the recipients. To our knowledge, this is the first demonstration of Bregs in response to cestodes, and we are unaware of any other study showing helminth-induced Bregs suppressing diseases associated with TH1, TH2, and TH1/2 immunity.

The induction of a Breg would be via contact with worm Ag or conditioning by the immune milieu, in which IL-4 and IL-13 are likely candidates given the stereotypic TH2 response to infection with helminths. B cells from naive mice exposed to H. diminuta extracts displayed no ability to suppress colitis. IL-4 plus IL-13 in vitro–educated B cells had a different regulatory program from HdBc(7d), being less effective in DNBS-induced colitis and largely ineffective in the oxazolone model.

Testing an in vivo requirement for IL-4 in the H. diminuta–elicited Breg, CD19+HdBc(7d) were isolated from infected IL-4Rα−/− and mast cell–deficient mice, with the former being incapable of responding to IL-4 and IL-13 and the latter being associated with deficient TH2 cytokine production following helminth infection (33). HdBc(7d) from these mice inhibited DNBS-induced colitis. It is noteworthy that Con A–stimulated splenocytes from mast cell–deficient mice produced levels of IL-10 similar to cells from WT mice (HdBc(7d) ablation of disease in mast cell deficient mice indicated that mast cell-B cell communication was not required for the suppression of DNBS-colitis). In addition, stimulated splenocytes from IL-4Rα−/− mice produced less IL-10 than those from WT mice did, suggesting that even in the presence of low amounts of IL-10, HdBc(7d) drive anti-colitic activity. Thus, despite a role for IL-10 in specific helminth-driven Bregs (22), H. diminuta suppression of DNBS colitis (25), and CFA monoarthritis (6), IL-10 is neither the key differentiation factor nor effector molecule in the HdBc(7d). Stimulated HdBc(7d) did not preferentially produce IL-10, and more conclusively, HdBc(7d) from IL-10−/− mice suppressed DNBS-induced colitis to a degree indistinguishable from cells from WT mice.

A small but significant increase in TGF-β was noted in HdBc(7d). TGF-β has been implicated in helminth-driven immunosuppression (43), and human B cells can produce TGF-β (44); therefore, it is tempting to speculate that the anti-colitic effect of HdBc(7d) may be due, in part, to TGF-β. The use of anti-TGFβ neutralizing Abs supports this supposition: disease in mice treated with DNBS plus HdBc(7d) plus anti–TGF-β was not statistically different from mice treated with only DNBS; however, mice treated with DNBS plus anti–TGF-β displayed exaggerated disease. Whereas TGF-β may be a component of the HdBc(7d) suppression of colitis the possibility that the anti–TGF-β worked by blocking recipient rather than donor cell TGF-β cannot be dismissed.

The interaction of H. diminuta, a gut lumen-dwelling parasite, with its host is different from that of abrasive nematodes and helminth species that cause tissue damage via migration through the host; this tissue damage has been posited as a driving force for anti-inflammatory events (45). Given this substantial difference in host–parasite interaction, the generation of a unique splenic CD19+ Breg in response to H. diminuta is plausible. Furthermore, descriptions of lymphocyte (including Bregs), innate adaptive lymphocytes, and macrophage subtypes (46, 47) attest to this possibility, supporting the contention that unique B cells could be mobilized in response to infection with cestode parasites.

A spectrum of Bregs have been identified based on expression of CD1d, CD5, CD23, FasL, Tim-1, and others (21, 44); for example, CD23hiCD5 and CD1dhiCD21hiCD23hi Bregs have been described in mice following infection with H. polygyrus and S. mansoni, respectively (22, 24). Whereas the total number of splenic CD19+/CD5+ cells was increased in infected mice, flow cytometry analyses did not reveal significant upregulation of canonical Breg markers on the HdBc(7d). Similarly, expression of NO (31) and ChAT (48), recently described components of B cell populations, were not distinguishing features of the H. diminuta–induced Bregs. Based on these results, we suggest that, analogous to B10 cells (49), the H. diminuta–induced Breg is defined by a functional program and not lineage specific markers. However, the H. diminuta–induced Breg did reside within the follicular zone of the spleen, and microarray analysis revealed a different signature in the HdBc(7d) compared with splenic CD19+ cells from naive mice. Some of the upregulated gene products may have anti-inflammatory or immunoregulatory function (e.g., complement component 1, q subcomponent [C1qb]) (50) and warrant further assessment. We speculate that transcriptome analysis with appropriate comparator groups (e.g., IL-4–treated B cells) could yield a HdBc(7d) signature for screening Bregs.

Temporal analysis of CFSE-labeled HdBc(7d) delivered to the peritoneal cavity revealed no obvious accumulation in the omentum, spleen, MLN or colon; substantial numbers remained in the peritoneal cavity. It is possible that small numbers of HdBc(7d) had trafficked to the colon or secondary lymphoid tissues and were below the level of confident detection by flow cytometry. The peritoneal cavity may be the inductive site for the anti-colitic activity of the HdBc(7d). In this context, it has been shown that NK cells delivered to the peritoneal cavity can remain there for 54 d (51), that macrophages in the peritoneal cavity can be important in propagating bystander nonspecific anti-tumor immune responses (52), and that peritoneal B10 B cells as immunotherapy can suppress DSS-induced colitis (49).

Mechanistically, Bregs can have intrinsic anti-inflammatory activity or could mobilize other immunoregulatory cells such as Foxp3+ Tregs (22). Foxp3 mRNA was not increased in mice receiving HdBc(7d), but this does not reflect function nor negate the possibility of recruitment of other regulatory T or B cells into the anti-colitic response. Although there may be differences in the mechanism of DNBS-induced colitis in WT and T cell–depleted mice, RAG1−/− mice display a DNBS phenotype and histopathology that are highly similar to WT mice. Suppression of DNBS-induced colitis in RAG1−/− mice that received HdBc(7d) revealed that recipient T and B cells were not required for the inhibition of colitis.

The data from RAG1−/− mice indicate either an intrinsic anti-colitic capacity in the HdBc(7d) or interaction with innate immune cells. We found that DNBS-induced colitis in RAG1−/− mice treated with clodronate and receiving HdBc(7d) was not different from mice treated with DNBS ± clodronate. Administration of clodronate-liposomes depletes phagocytes, predominately MØ (20, 53) (confirmed by F4/80 staining), indicating B cell–MØ communication in the amelioration of colitis. There are few data on B cell–MØ cross-talk, but it is not unprecedented; peritoneal MØ from B cell–deficient mice produced more TNF-α and IL-1β in response to LPS (54), and monocyte recruitment in murine myocardial infarction was triggered by B cells (55). The current study demonstrates B cell–MØ cooperation in the suppression of colitis. Alternatively activated macrophages can block colitis (20), but the induction of AAMs is unlikely to underlie the HdBc(7d) anti-colitic effect because qPCR revealed no increase in arginase, Ym1, and Relmα mRNA expression in MØ cocultured with HdBc(7d) or colonic extracts of mice given HdBc(7d). Indeed, an anti-inflammatory MØ that was Ym1/Relmα was implicated in the suppression of DSS-induced colitis following infection with S. mansoni (18). Coculture of HdBc(7d) with bone marrow-derived MØ resulted in increased LPS-induced IL-10 production by the MØ. So whereas HdBc(7d) appear to be neither a target nor a source of IL-10, their induction of MØ-derived IL-10 would contribute to the suppression of colitis, which is compatible with the role for IL-10 in the H. diminuta suppression of DNBS-induced colitis (25).

Finally, the reciprocal MØ–B cell communication was considered. Coculture of HdBc(7d) with MØ or CAMs resulted in upregulation of CD1d and PDL1 on the B cell. The former would allow engagement of NK T cells, and the latter could give the B cell an additional anti-colitic strategy as described for PDL1 and the anergy of T cells following infection with S. mansoni (56). As the mechanism of HdBc(7d) action is unraveled, it will be important to dissect MØ–B cell communication and the consequences of activation of each cell as elements of the suppression of colitis.

A complex web of immunity arises following infection with helminth parasites, driven by the opposing forces of the host’s efforts to eliminate the parasite and the parasite’s attempts to circumvent the host’s anti-worm response. In this study, we identify a putatively unique splenic follicular zone CD19+ B cell mobilized in response to H. diminuta that requires neither IL-4 nor IL-10 for its differentiation and does not overexpress typical markers of Bregs (i.e., CD1d, CD23) or IL-10, and whose anti-colitic activity can be independent of T and B cells. Rather, it appears that this H. diminuta CD19+ Breg is a source of TGF-β and exerts an anti-colitic effect by cooperating with MØ whose immunoregulatory properties may be affected by TGF-β and other factors that await identification or cognate interaction with the HdBc(7d). Precise characterization of the phenotype and mechanism of action of this CD19+HdBc(7d) coupled with the ability to reprogram human B cells into such a phenotype could be a valuable addition to the treatments available for mucosal inflammatory disease.

We thank Dr. Hermelijn Smits (Leiden University Medical Center, Leiden, the Netherlands) for helpful discussions, Liselotte Lenner (Faculty of Health Sciences, Linköping University, Sweden) for performing microarray hybridizations, Dr. Xiuling Wang (Faculty of Medicine, University of Calgary) for assistance with microarray analysis, and Laurie Kennedy and Yiping Liu (University of Calgary Flow Cytometry facilities) for help during cell sorting experiments. N.v.R. provided the liposomes.

This work was funded by a Discovery Grant from the Natural Sciences and Engineering Research Council of Canada (to D.M.M.) and in part by the Crohn’s and Colitis Foundation of Canada. J.L.R. is a recipient of postdoctoral fellowships from Alberta Innovates-Health Solutions, the Canadian Institutes for Health Research, the Canadian Association of Gastroenterology, and Janssen Pharmaceuticals. M.R.F. is a recipient of Graduate Studentships from the Canadian Institutes for Health Research, the Canadian Digestive Health Foundation, and Alberta Innovates-Health Solutions. D.M.M. holds a Canada Research Chair (Tier 1) in Intestinal Immunophysiology. J.L.R. and D.M.M. are supported by the Host–Parasite Interactions Natural Sciences and Engineering Research Council Collaborative Research and Training Experience Programme.

The online version of this article contains supplemental material.

Abbreviations used in this article:

AAM

alternatively activated macrophage

Bc

splenic B cell purified from uninfected mice

BcIL-4/13

splenic B cell from naive mice exposed to IL-4 and IL-13

Breg

regulatory B cell

CAM

classically activated macrophage

ChAT

choline acetyltransferase

clodL

clodronate-loaded liposome

C1qb

complement component 1, q subcomponent

DNBS

dinitrobenzene sulfonic acid

DSS

dextran sodium sulphate

HdAg

PBS-soluble crude Ag extract of adult Hymenolepis diminuta

HdBc(3d)

(7d), (14d), splenic B cell purified from mice infected with H. diminuta 3, 7, and 14 d previously

i.r.

intrarectal

KO

knockout

macrophage

MLN

mesenteric lymph node

MPO

myeloperoxidase

PBSL

PBS-loaded liposome

PDL1

programmed death ligand 1

qPCR

quantitative PCR

Relmα

resistin-like molecule α

TH2

T 2 helper

Treg

regulatory T cell

WT

wild-type

Ym1

murine chitinase 3–like protein.

1
Maizels
R. M.
,
Yazdanbakhsh
M.
.
2003
.
Immune regulation by helminth parasites: cellular and molecular mechanisms.
Nat. Rev. Immunol.
3
:
733
744
.
2
McSorley
H. J.
,
Maizels
R. M.
.
2012
.
Helminth infections and host immune regulation.
Clin. Microbiol. Rev.
25
:
585
608
.
3
Anthony
R. M.
,
Rutitzky
L. I.
,
Urban
J. F.
 Jr.
,
Stadecker
M. J.
,
Gause
W. C.
.
2007
.
Protective immune mechanisms in helminth infection.
Nat. Rev. Immunol.
7
:
975
987
.
4
Mishra
P. K.
,
Patel
N.
,
Wu
W.
,
Bleich
D.
,
Gause
W. C.
.
2013
.
Prevention of type 1 diabetes through infection with an intestinal nematode parasite requires IL-10 in the absence of a Th2-type response.
Mucosal Immunol.
6
:
297
308
.
5
Cooke
A.
,
Tonks
P.
,
Jones
F. M.
,
O’Shea
H.
,
Hutchings
P.
,
Fulford
A. J.
,
Dunne
D. W.
.
1999
.
Infection with Schistosoma mansoni prevents insulin dependent diabetes mellitus in non-obese diabetic mice.
Parasite Immunol.
21
:
169
176
.
6
Shi
M.
,
Wang
A.
,
Prescott
D.
,
Waterhouse
C. C.
,
Zhang
S.
,
McDougall
J. J.
,
Sharkey
K. A.
,
McKay
D. M.
.
2011
.
Infection with an intestinal helminth parasite reduces Freund’s complete adjuvant-induced monoarthritis in mice.
Arthritis Rheum.
63
:
434
444
.
7
Reyes
J. L.
,
Espinoza-Jiménez
A. F.
,
González
M. I.
,
Verdin
L.
,
Terrazas
L. I.
.
2011
.
Taenia crassiceps infection abrogates experimental autoimmune encephalomyelitis.
Cell. Immunol.
267
:
77
87
.
8
Wilson
M. S.
,
Taylor
M. D.
,
Balic
A.
,
Finney
C. A.
,
Lamb
J. R.
,
Maizels
R. M.
.
2005
.
Suppression of allergic airway inflammation by helminth-induced regulatory T cells.
J. Exp. Med.
202
:
1199
1212
.
9
Bashir
M. E.
,
Andersen
P.
,
Fuss
I. J.
,
Shi
H. N.
,
Nagler-Anderson
C.
.
2002
.
An enteric helminth infection protects against an allergic response to dietary antigen.
J. Immunol.
169
:
3284
3292
.
10
Hernandez
J. L.
,
Leung
G.
,
McKay
D. M.
.
2013
.
Cestode regulation of inflammation and inflammatory diseases.
Int. J. Parasitol.
43
:
233
243
.
11
Correale
J.
,
Farez
M. F.
.
2011
.
The impact of parasite infections on the course of multiple sclerosis.
J. Neuroimmunol.
233
:
6
11
.
12
Reyes
J. L.
,
Terrazas
L. I.
.
2007
.
The divergent roles of alternatively activated macrophages in helminthic infections.
Parasite Immunol.
29
:
609
619
.
13
McSorley
H. J.
,
Harcus
Y. M.
,
Murray
J.
,
Taylor
M. D.
,
Maizels
R. M.
.
2008
.
Expansion of Foxp3+ regulatory T cells in mice infected with the filarial parasite Brugia malayi.
J. Immunol.
181
:
6456
6466
.
14
Hang
L.
,
Blum
A. M.
,
Setiawan
T.
,
Urban
J. P.
 Jr.
,
Stoyanoff
K. M.
,
Weinstock
J. V.
.
2013
.
Heligmosomoides polygyrus bakeri infection activates colonic Foxp3+ T cells enhancing their capacity to prevent colitis.
J. Immunol.
191
:
1927
1934
.
15
van der Vlugt
L. E.
,
Labuda
L. A.
,
Ozir-Fazalalikhan
A.
,
Lievers
E.
,
Gloudemans
A. K.
,
Liu
K. Y.
,
Barr
T. A.
,
Sparwasser
T.
,
Boon
L.
,
Ngoa
U. A.
, et al
.
2012
.
Schistosomes induce regulatory features in human and mouse CD1d(hi) B cells: inhibition of allergic inflammation by IL-10 and regulatory T cells.
PLoS ONE
7
:
e30883
.
16
Elliott
D. E.
,
Setiawan
T.
,
Metwali
A.
,
Blum
A.
,
Urban
J. F.
 Jr.
,
Weinstock
J. V.
.
2004
.
Heligmosomoides polygyrus inhibits established colitis in IL-10-deficient mice.
Eur. J. Immunol.
34
:
2690
2698
.
17
Blum
A. M.
,
Hang
L.
,
Setiawan
T.
,
Urban
J. P.
 Jr.
,
Stoyanoff
K. M.
,
Leung
J.
,
Weinstock
J. V.
.
2012
.
Heligmosomoides polygyrus bakeri induces tolerogenic dendritic cells that block colitis and prevent antigen-specific gut T cell responses.
J. Immunol.
189
:
2512
2520
.
18
Smith
P.
,
Mangan
N. E.
,
Walsh
C. M.
,
Fallon
R. E.
,
McKenzie
A. N.
,
van Rooijen
N.
,
Fallon
P. G.
.
2007
.
Infection with a helminth parasite prevents experimental colitis via a macrophage-mediated mechanism.
J. Immunol.
178
:
4557
4566
.
19
McKay
D. M.
2010
.
The immune response to and immunomodulation by Hymenolepis diminuta.
Parasitology
137
:
385
394
.
20
Hunter
M. M.
,
Wang
A.
,
Parhar
K. S.
,
Johnston
M. J.
,
Van Rooijen
N.
,
Beck
P. L.
,
McKay
D. M.
.
2010
.
In vitro-derived alternatively activated macrophages reduce colonic inflammation in mice.
Gastroenterology
138
:
1395
1405
.
21
Klinker
M. W.
,
Lundy
S. K.
.
2012
.
Multiple mechanisms of immune suppression by B lymphocytes.
Mol. Med.
18
:
123
137
.
22
Amu
S.
,
Saunders
S. P.
,
Kronenberg
M.
,
Mangan
N. E.
,
Atzberger
A.
,
Fallon
P. G.
.
2010
.
Regulatory B cells prevent and reverse allergic airway inflammation via FoxP3-positive T regulatory cells in a murine model.
J. Allergy Clin. Immunol.
125
:
1114
1124, e8
.
23
Hussaarts
L.
,
van der Vlugt
L. E.
,
Yazdanbakhsh
M.
,
Smits
H. H.
.
2011
.
Regulatory B-cell induction by helminths: implications for allergic disease.
J. Allergy Clin. Immunol.
128
:
733
739
.
24
Wilson
M. S.
,
Taylor
M. D.
,
O’Gorman
M. T.
,
Balic
A.
,
Barr
T. A.
,
Filbey
K.
,
Anderton
S. M.
,
Maizels
R. M.
.
2010
.
Helminth-induced CD19+CD23hi B cells modulate experimental allergic and autoimmune inflammation.
Eur. J. Immunol.
40
:
1682
1696
.
25
Hunter
M. M.
,
Wang
A.
,
Hirota
C. L.
,
McKay
D. M.
.
2005
.
Neutralizing anti-IL-10 antibody blocks the protective effect of tapeworm infection in a murine model of chemically induced colitis.
J. Immunol.
174
:
7368
7375
.
26
Johnston
M. J.
,
Wang
A.
,
Catarino
M. E.
,
Ball
L.
,
Phan
V. C.
,
MacDonald
J. A.
,
McKay
D. M.
.
2010
.
Extracts of the rat tapeworm, Hymenolepis diminuta, suppress macrophage activation in vitro and alleviate chemically induced colitis in mice.
Infect. Immun.
78
:
1364
1375
.
27
DeKoter
R. P.
,
Geadah
M.
,
Khoosal
S.
,
Xu
L. S.
,
Thillainadesan
G.
,
Torchia
J.
,
Chin
S. S.
,
Garrett-Sinha
L. A.
.
2010
.
Regulation of follicular B cell differentiation by the related E26 transformation-specific transcription factors PU.1, Spi-B, and Spi-C.
J. Immunol.
185
:
7374
7384
.
28
Wang
A.
,
Fernando
M.
,
Leung
G.
,
Phan
V.
,
Smyth
D.
,
McKay
D. M.
.
2010
.
Exacerbation of oxazolone colitis by infection with the helminth Hymenolepis diminuta: involvement of IL-5 and eosinophils.
Am. J. Pathol.
177
:
2850
2859
.
29
Ding
Q.
,
Yeung
M.
,
Camirand
G.
,
Zeng
Q.
,
Akiba
H.
,
Yagita
H.
,
Chalasani
G.
,
Sayegh
M. H.
,
Najafian
N.
,
Rothstein
D. M.
.
2011
.
Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice.
J. Clin. Invest.
121
:
3645
3656
.
30
Leung
G.
,
Wang
A.
,
Fernando
M.
,
Phan
V. C.
,
McKay
D. M.
.
2013
.
Bone marrow-derived alternatively activated macrophages reduce colitis without promoting fibrosis: participation of IL-10.
Am. J. Physiol. Gastrointest. Liver Physiol.
304
:
G781
G792
.
31
Fritz
J. H.
,
Rojas
O. L.
,
Simard
N.
,
McCarthy
D. D.
,
Hapfelmeier
S.
,
Rubino
S.
,
Robertson
S. J.
,
Larijani
M.
,
Gosselin
J.
,
Ivanov
I. I.
, et al
.
2012
.
Acquisition of a multifunctional IgA+ plasma cell phenotype in the gut.
Nature
481
:
199
203
.
32
Underhill
G. H.
,
Kolli
K. P.
,
Kansas
G. S.
.
2003
.
Complexity within the plasma cell compartment of mice deficient in both E- and P-selectin: implications for plasma cell differentiation.
Blood
102
:
4076
4083
.
33
Hepworth
M. R.
,
Daniłowicz-Luebert
E.
,
Rausch
S.
,
Metz
M.
,
Klotz
C.
,
Maurer
M.
,
Hartmann
S.
.
2012
.
Mast cells orchestrate type 2 immunity to helminths through regulation of tissue-derived cytokines.
Proc. Natl. Acad. Sci. USA
109
:
6644
6649
.
34
Saleem
S. J.
,
Martin
R. K.
,
Morales
J. K.
,
Sturgill
J. L.
,
Gibb
D. R.
,
Graham
L.
,
Bear
H. D.
,
Manjili
M. H.
,
Ryan
J. J.
,
Conrad
D. H.
.
2012
.
Cutting edge: mast cells critically augment myeloid-derived suppressor cell activity.
J. Immunol.
189
:
511
515
.
35
Duan
L.
,
Chen
J.
,
Zhang
H.
,
Yang
H.
,
Zhu
P.
,
Xiong
A.
,
Xia
Q.
,
Zheng
F.
,
Tan
Z.
,
Gong
F.
,
Fang
M.
.
2012
.
Interleukin-33 ameliorates experimental colitis through promoting Th2/Foxp3⁺ regulatory T-cell responses in mice.
Mol. Med.
18
:
753
761
.
36
Boehm
F.
,
Martin
M.
,
Kesselring
R.
,
Schiechl
G.
,
Geissler
E. K.
,
Schlitt
H. J.
,
Fichtner-Feigl
S.
.
2012
.
Deletion of Foxp3+ regulatory T cells in genetically targeted mice supports development of intestinal inflammation.
BMC Gastroenterol.
12
:
97
.
37
Dutra
R. C.
,
Cola
M.
,
Leite
D. F.
,
Bento
A. F.
,
Claudino
R. F.
,
Nascimento
A. F.
,
Leal
P. C.
,
Calixto
J. B.
.
2011
.
Inhibitor of PI3Kγ ameliorates TNBS-induced colitis in mice by affecting the functional activity of CD4+CD25+FoxP3+ regulatory T cells.
Br. J. Pharmacol.
163
:
358
374
.
38
Carter
N. A.
,
Rosser
E. C.
,
Mauri
C.
.
2012
.
Interleukin-10 produced by B cells is crucial for the suppression of Th17/Th1 responses, induction of T regulatory type 1 cells and reduction of collagen-induced arthritis.
Arthritis Res. Ther.
14
:
R32
R39
.
39
Ray
A.
,
Basu
S.
,
Williams
C. B.
,
Salzman
N. H.
,
Dittel
B. N.
.
2012
.
A novel IL-10-independent regulatory role for B cells in suppressing autoimmunity by maintenance of regulatory T cells via GITR ligand.
J. Immunol.
188
:
3188
3198
.
40
Ferreira
I.
,
Smyth
D.
,
Gaze
S.
,
Aziz
A.
,
Giacomin
P.
,
Ruyssers
N.
,
Artis
D.
,
Laha
T.
,
Navarro
S.
,
Loukas
A.
,
McSorley
H. J.
.
2013
.
Hookworm excretory/secretory products induce interleukin-4 (IL-4)+ IL-10+ CD4+ T cell responses and suppress pathology in a mouse model of colitis.
Infect. Immun.
81
:
2104
2111
.
41
Schnoeller
C.
,
Rausch
S.
,
Pillai
S.
,
Avagyan
A.
,
Wittig
B. M.
,
Loddenkemper
C.
,
Hamann
A.
,
Hamelmann
E.
,
Lucius
R.
,
Hartmann
S.
.
2008
.
A helminth immunomodulator reduces allergic and inflammatory responses by induction of IL-10-producing macrophages.
J. Immunol.
180
:
4265
4272
.
42
Yanaba
K.
,
Yoshizaki
A.
,
Asano
Y.
,
Kadono
T.
,
Tedder
T. F.
,
Sato
S.
.
2011
.
IL-10-producing regulatory B10 cells inhibit intestinal injury in a mouse model.
Am. J. Pathol.
178
:
735
743
.
43
Grainger
J. R.
,
Smith
K. A.
,
Hewitson
J. P.
,
McSorley
H. J.
,
Harcus
Y.
,
Filbey
K. J.
,
Finney
C. A.
,
Greenwood
E. J.
,
Knox
D. P.
,
Wilson
M. S.
, et al
.
2010
.
Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway.
J. Exp. Med.
207
:
2331
2341
.
44
Vadasz
Z.
,
Haj
T.
,
Kessel
A.
,
Toubi
E.
.
2013
.
B-regulatory cells in autoimmunity and immune mediated inflammation.
FEBS Lett.
587
:
2074
2078
.
45
Gause
W. C.
,
Wynn
T. A.
,
Allen
J. E.
.
2013
.
Type 2 immunity and wound healing: evolutionary refinement of adaptive immunity by helminths.
Nat. Rev. Immunol.
13
:
607
614
.
46
Fuchs
A.
,
Vermi
W.
,
Lee
J. S.
,
Lonardi
S.
,
Gilfillan
S.
,
Newberry
R. D.
,
Cella
M.
,
Colonna
M.
.
2013
.
Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-γ-producing cells.
Immunity
38
:
769
781
.
47
Gordon
S.
,
Mantovani
A.
.
2011
.
Diversity and plasticity of mononuclear phagocytes.
Eur. J. Immunol.
41
:
2470
2472
.
48
Reardon
C.
,
Duncan
G. S.
,
Brüstle
A.
,
Brenner
D.
,
Tusche
M. W.
,
Olofsson
P. S.
,
Rosas-Ballina
M.
,
Tracey
K. J.
,
Mak
T. W.
.
2013
.
Lymphocyte-derived ACh regulates local innate but not adaptive immunity.
Proc. Natl. Acad. Sci. USA
110
:
1410
1415
.
49
Maseda
D.
,
Candando
K. M.
,
Smith
S. H.
,
Kalampokis
I.
,
Weaver
C. T.
,
Plevy
S. E.
,
Poe
J. C.
,
Tedder
T. F.
.
2013
.
Peritoneal cavity regulatory B cells (B10 cells) modulate IFN-γ+CD4+ T cell numbers during colitis development in mice.
J. Immunol.
191
:
2780
2795
.
50
Xiao
Y.
,
Xu
J.
,
Wang
S.
,
Mao
C.
,
Jin
M.
,
Ning
G.
,
Xu
J.
,
Zhang
Y.
.
2010
.
Genetic ablation of steroid receptor coactivator-3 promotes PPAR-β-mediated alternative activation of microglia in experimental autoimmune encephalomyelitis.
Glia
58
:
932
942
.
51
Geller
M. A.
,
Knorr
D. A.
,
Hermanson
D. A.
,
Pribyl
L.
,
Bendzick
L.
,
McCullar
V.
,
Miller
J. S.
,
Kaufman
D. S.
.
2013
.
Intraperitoneal delivery of human natural killer cells for treatment of ovarian cancer in a mouse xenograft model.
Cytotherapy
15
:
1297
1306
.
52
Sedlacek
A. L.
,
Gerber
S. A.
,
Randall
T. D.
,
van Rooijen
N.
,
Frelinger
J. G.
,
Lord
E. M.
.
2013
.
Generation of a dual-functioning antitumor immune response in the peritoneal cavity.
Am. J. Pathol.
183
:
1318
1328
.
53
Zhao
A.
,
Urban
J. F.
 Jr.
,
Anthony
R. M.
,
Sun
R.
,
Stiltz
J.
,
van Rooijen
N.
,
Wynn
T. A.
,
Gause
W. C.
,
Shea-Donohue
T.
.
2008
.
Th2 cytokine-induced alterations in intestinal smooth muscle function depend on alternatively activated macrophages.
Gastroenterology
135
:
217
225, e1
.
54
Wong
S. C.
,
Puaux
A. L.
,
Chittezhath
M.
,
Shalova
I.
,
Kajiji
T. S.
,
Wang
X.
,
Abastado
J. P.
,
Lam
K. P.
,
Biswas
S. K.
.
2010
.
Macrophage polarization to a unique phenotype driven by B cells.
Eur. J. Immunol.
40
:
2296
2307
.
55
Zouggari
Y.
,
Ait-Oufella
H.
,
Bonnin
P.
,
Simon
T.
,
Sage
A. P.
,
Guérin
C.
,
Vilar
J.
,
Caligiuri
G.
,
Tsiantoulas
D.
,
Laurans
L.
, et al
.
2013
.
B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction.
Nat. Med.
19
:
1273
1280
.
56
Smith
P.
,
Walsh
C. M.
,
Mangan
N. E.
,
Fallon
R. E.
,
Sayers
J. R.
,
McKenzie
A. N.
,
Fallon
P. G.
.
2004
.
Schistosoma mansoni worms induce anergy of T cells via selective up-regulation of programmed death ligand 1 on macrophages.
J. Immunol.
173
:
1240
1248
.

The authors have no financial conflicts of interest.

Supplementary data