Skip to main content
Advertisement
  • Loading metrics

IL-22 Restrains Tapeworm-Mediated Protection against Experimental Colitis via Regulation of IL-25 Expression

  • José L. Reyes ,

    jlreyesh@ucalgary.ca (JLR); dmckay@ucalgary.ca (DMM)

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

  • Maria R. Fernando,

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

  • Fernando Lopes,

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

  • Gabriella Leung,

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

  • Nicole L. Mancini,

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

  • Chelsea E. Matisz,

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

  • Arthur Wang,

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

  • Derek M. McKay

    jlreyesh@ucalgary.ca (JLR); dmckay@ucalgary.ca (DMM)

    Affiliation Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada

Abstract

Interleukin (IL)-22, an immune cell-derived cytokine whose receptor expression is restricted to non-immune cells (e.g. epithelial cells), can be anti-inflammatory and pro-inflammatory. Mice infected with the tapeworm Hymenolepis diminuta are protected from dinitrobenzene sulphonic acid (DNBS)-induced colitis. Here we assessed expulsion of H. diminuta, the concomitant immune response and the outcome of DNBS-induced colitis in wild-type (WT) and IL-22 deficient mice (IL-22-/-) ± infection. Interleukin-22-/- mice had a mildly impaired ability to expel the worm and this correlated with reduced or delayed induction of TH2 immunity as measured by splenic and mesenteric lymph node production of IL-4, IL-5 and IL-13 and intestinal Muc-2 mRNA and goblet cell hyperplasia; in contrast, IL-25 increased in the small intestine of IL-22-/- mice 8 and 12 days post-infection compared to WT mice. In vitro experiments revealed that H. diminuta directly evoked epithelial production of IL-25 that was inhibited by recombinant IL-22. Also, IL-10 and markers of regulatory T cells were increased in IL-22-/- mice that displayed less DNBS (3 mg, ir. 72h)-induced colitis. Wild-type mice infected with H. diminuta were protected from colitis, as were infected IL-22-/- mice and the latter to a degree that they were almost indistinguishable from control, non-DNBS treated mice. Finally, treatment with anti-IL-25 antibodies exaggerated DNBS-induced colitis in IL-22-/- mice and blocked the anti-colitic effect of infection with H. diminuta. Thus, IL-22 is identified as an endogenous brake on helminth-elicited TH2 immunity, reducing the efficacy of expulsion of H. diminuta and limiting the effectiveness of the anti-colitic events mobilized following infection with H. diminuta in a non-permissive host.

Author Summary

Interleukin (IL)-22, produced by innate and adaptive immune cells, plays a complex role in immunity; under specific conditions, targeting this cytokine could treat inflammatory diseases. The hygiene hypothesis suggests infection with helminth parasites could ameliorate inflammation. Here we show that IL-22 is required to activate early events (i.e. type 2 cytokines and mucin expression) in the response to the non-invasive cestode Hymenolepis diminuta. Strikingly, expression of regulatory factors (IL-10, IL-25, Foxp3), which arise following H. diminuta infection, were either enhanced or sustained in IL-22-/- mice, uncovering a novel role for IL-22 as a brake for these regulatory events following infection with this parasitic helminth. Moreover, DNBS-induced colitis was significantly less severe in IL-22-/- compared to wild-type mice: IL-22-/- mice infected with H. dimunta 8-days prior to the induction of colitis had negligible disease. Immunoneutralization of IL-25 exaggerated DNBS-induced colitis in the IL-22-/- mice and ablated the anti-colitic effect of infection with H. diminta. Thus, while immune events in the early response to infection with H. diminuta are delayed in IL-22-/- mice (as is worm expulsion), the compensatory enhancement of IL-25 (and other immunoregulatory elements (e.g. IL-10)) provide resistance to colitis and also promote the anti-colitic effect driven as a consequence of the response to infection with H. diminuta. The data confirm the complex role of IL-22 in intestinal immunity.

Introduction

Interleukin (IL)-22, a member of the IL-10 family, is produced predominantly by innate (NK cells (NK22), γδ T cells, innate lymphoid cells type 3 (ILC3s) and adaptive (CD4+ Th22 and Th17, CD8+ T cells) immune cells: a non-immune source of IL-22 has not been described. The heterodimeric IL-22 receptor consists of the IL-10R2 subunit and the unique IL-22R1 subunit, and is restricted to non-hematopoetic cells (e.g. hepatocytes and epithelium of the gastrointestinal tract) [1]. Thus, IL-22 is an immune cell-derived mediator that acts exclusively on non-immune cells and as such is an attractive target for therapeutic intervention [2, 3].

Data from studies of the gastrointestinal tract suggest that the role of IL-22 is contextual, with beneficial or detrimental affects depending on the nature of the disease or immune activity being assessed. For example, IL-22-/- mice are more susceptible to colitis induced by dextran sodium sulfate (DSS) [4] and retinoic acid suppression of DSS-induced colitis was associated with increased IL-22 [5]. Similarly, local delivery of the IL-22 gene attenuated the spontaneous colitis that develops in T cell receptor (TCR)-α knockout (KO) mice [6] and that evoked by transfer of naïve CD45RBhi T cells into RAG2-/- mice [4]. However, IL-22, mobilized by IL-23, was implicated in the exaggeration of murine colitis induced by anti-CD40 activation in RAG1-/- mice [7]. Fewer IL-22+ cells have been described in inflamed tissue from patients with ulcerative colitis compared to healthy individuals [8]. In contrast, ILC3 from patients with mild-moderate ulcerative colitis were reported to have increased IL-22 production [9].

This duality of IL-22 function extends beyond the gut. Interleukin-22 can promote hepatocyte survival in acute mouse models of liver damage [10], while IL-22 recruitment of Th17 cells has been implicated in chronic liver inflammation of hepatitis B-infected individuals [11]. Pro- and anti-inflammatory roles have been described for IL-22 in murine models of arthritis [12]; for example, IL-22 was implicated in the enhancement or suppression of collagen-induced arthritis in mice co-treated with the parasitic nematode-derived molecule, ES-62 [13].

The role of IL-22 following infection is equally diverse, where it has been shown to protect mice from infection with Citrobacter rodentium and Salmonella enterica [14], but appears not to affect the outcome of infection with Mycobacterium avium [15]; susceptibility to Salmonella has been reported [16]. The route of pathogen entry into the body can be important, IL-22 acting downstream of IL-23, promoted resistance against intragastrically or intravenously delivered Candida albicans [17], but played no role in the response to cutaneous C. albicans [18]. Two independent studies demonstrated roles for IL-22 in the intestinal pathophysiology associated with infection with Toxoplasma gondii [15, 19]. With respect to infection with helminth parasites, Wilson et. al. found no role for IL-22 in the murine response to Schistosoma mansoni [15], whereas goblet cell hyperplasia and mucin secretion, a key effector in the gut, was driven by IL-22 following infection with nematodes [20]. Increased IL-22 has been demonstrated in individuals with established hookworm infection although its function was not defined [21]. A report of self-infection with the nematode parasite Trichuris trichiura to treat ulcerative colitis documented increased numbers of CD4+IL22+ cells [22].

Infection with the rat tapeworm, Hymenolepis diminuta, protects mice from colitis induced by intra-rectal (i.r.) instillation of the haptenizing agent, 2,4-dinitrobenzene sulphonic acid (DNBS) [23]. Given the pivotal role that IL-22 can play in immune-stromal cell communication and the disparite data on this cytokine in the response to infection (and general lack of data in relation to helminths) and regulation of inflammation, the current study assessed the impact of the absence of IL-22 in (1) the expulsion of H. diminuta from its non-permissive mouse host and the concomitant immune response, and (2) whether the anti-colitic effect of infection with H. diminuta was modified.

Results and Discussion

IL-22-/- mice display defective expulsion of H. diminuta and reduced early TH2 response

The role of IL-22 in modifying the host response to infection with helminth parasites appears to be determined by the nature of the infection. For example, worm burden and granuloma size is not different in schistosoma-infected WT and IL-22-/- mice [15], whereas IL-22 was important in the goblet cell hyperplasia and mucin secretion response following infection with the intestinal nematodes, Trichuris muris and Nippostrongylus brasiliensis [20]. The tapeworm H. diminuta is unique amongst helminths that infect the intestine as it does negligible, if any, damage to the host: it lacks a tissue migratory phase and the absence of hooks on the scolex means it is not abrasive. IL-22-/- mice displayed a slight delay in the kinetics of expulsion of H. diminuta: only 22% (2/9 mice) of infected IL-22-/- mice had expelled H. diminuta by 8 days post-infection (dpi) compared to 55% (5/9 mice) of WT mice (Fig 1); at this time-point 33% of infected IL-22-/- mice harboured 3 or 4 worms, burdens not observed in WT mice. At 12 dpi, H. diminuta had been completed expelled from WT and IL-22-/- mice, suggesting that while IL-22 signaling promotes a rapid anti-H. diminuta response the duration of infection is not prolonged in the absence of this cytokine.

thumbnail
Fig 1. Absence of IL-22 alters the expulsion kinetics of H. diminuta from mice.

Wild-type (WT; gray symbols) and IL-22-/- (black symbols) mice were orally infected with 5 H. diminuta cysticrecoids and at the indicated time-points were euthanized, the small intestine removed, flushed with PBS and retrieved parasites counted (lines indicate mean ± SEM; n = 6–9 at each point from 3 independent experiments; *p <0.05 by student’s t test).

https://doi.org/10.1371/journal.ppat.1005481.g001

Mobilization of TH2-type cytokines (i.e. IL-4, IL-5 and IL-13) is a hallmark of the immune response following infection with parasitic helminths [24]. Consistent with previous findings [25], mitogen stimulation of splenocytes or mesenteric lymph node (MLN) cells from WT mice resulted in increased IL-4, IL-5 and IL-13 by 4-dpi (Fig 2A and 2B), declining to control levels by 12-dpi. Time-matched analyses revealed reduced levels of the 3 cytokines from MLN and spleen of IL-22-/- mice on day 4-dpi compared to WT mice, that rebounded to match or exceed those of WT mice by day 8-pdi (the exception being IL-13 production by MLN cells) (Fig 2A and 2B). By 12-dpi there were no differences in splenic and MLN-derived IL-4, IL-5 or IL-13 in infected WT and IL-22-/- mice. Measurement of the TH1 cytokine IFN-γ from conA-stimulated splenocytes revealed no differences between WT and IL-22-/- mice over the 12-day infection period (S1 Fig). In addition, qPCR revealed reduced expression of IL-4, IL-10 and IL-25 mRNA in intestinal tissue from infected IL-22-/- mice compared to WT animals at 4-dpi, with a rebound heightened expression in all 3 cyokines by 8-dpi, which unlike the spleen and MLN was extended until 12-dpi (Fig 2C) (end of experiment). This delay in the production of key TH2 effector cytokines parallels the delay in expulsion of H. diminuta from IL-22-/- mice and the events are likely to be causally linked. These data align with the requirement for IL-25 in the expulsion of nematode parasites from mice [2628]. Fascinatingly, and in accordance with IL-22’s dual functions [13], the diminished TH2 responses in IL-22-/- H. diminuta-infected mice suggests an important role for innate immunity early in the response to helminths and additional studies are needed to precisely define this.

thumbnail
Fig 2. IL-22 absence results in altered TH2 immune responses.

Wild-type (WT; gray bars) and IL-22-/- (black bars) mice were infected with 5 H. diminuta cysticrecoids and at indicated time-points were euthanized and spleen (A), mesenteric lymph node (MLN) (B) and portions of mid-small intestine (C) were excised. Spleen and MLN cell suspensions were generated and incubated for 48 hr with conA (5 μg/ml) and TH2 cytokines quantified by ELISA. Intestinal tissue was assessed by qPCR (data are mean ± SEM; n = 9 from 3 independent experiments; * and #, p <0.05 compared to control and time-matched WT, respectively).

https://doi.org/10.1371/journal.ppat.1005481.g002

Interleukin-4 has been implicated in the regulation of goblet cell hyperplasia following infection with helminth parasites [29]. Indeed, mucin synthesis and release are important, often critical, effector responses against enteric helminths [30] and goblet cell hyperplasia follows the kinetics of H. diminuta expulsion from WT mice [31]. Four dpi mRNA for the secreted mucin, Muc-2, was increased in the small intestine of infected WT and to a lesser extent in IL-22-/- mice: and while Muc-2 mRNA expression declined in the intestine of WT mice, in IL-22-/- mice the elevated Muc-2 expression was maintained at 8-dpi, paralleling the kinetics of H. diminuta expulsion (Fig 3A). The Muc-1 gene encodes a transmembrane bound mucin; little is known of its function [32]. Muc-1 mRNA was significantly upregulated in H. diminuta-infected IL-22-/- mice at 8- and 12-dpi and it is tempting to speculate that this might compensate for the reduced Muc-2 signal at 4-dpi in these mice (Fig 3A). Rats, the natural definitive host for H. diminuta, infected with 5 cysticercoids show no increase in Muc-2 mRNA, whereas a 50 cysticercoid oral inoculum resulted in increased Muc-2 mRNA, and ≤15 worms established in the gut [33].

thumbnail
Fig 3. Mice lacking IL-22 display reduced expression of mucin mRNA and fewer goblet cells at early time-points following infection with H. diminuta.

Small intestine from WT (gray bars) and IL-22-/- (black bars) mice was flushed with sterile PBS and mRNA extracted from mid-small intestine and used to analyze Muc-1 and Muc-2 mRNA by qPCR (A) (target gene normalised against RNA 18s housekeeping gene). Goblet cells were identified by periodic acid-Schiff staining of sections on coded slides (B, C) and enumerated microscopically base on intact villus-crypt units (VCU) (data are mean ± SEM; n = 6–9 from 3 experiments, * and #, p <0.05 compared to control and time-matched WT, respectively).

https://doi.org/10.1371/journal.ppat.1005481.g003

Histochemical staining revealed increased numbers of mucus-containing goblet cells in the small intestine of H. diminuta-infected WT mice (Fig 3B) [31]; however, intestine from infected IL-22-/- mice displayed no significant increase in goblet cells at 4-dpi (Fig 3B). The reduced Muc-2 expression and parallel changes in goblet cell numbers in IL-22-/- mice could contribute to the increased worm burden observed at 8-dpi, while maintenance of the Muc-2 signal and sustained goblet cell numbers (Fig 3C) may allow for these mice to catch-up with WT animals, fully expelling H. diminuta by 12-dpi. However, neither Muc2 mRNA nor goblet cell numbers are substantially increased beyond WT levels at 12-dpi, despite increased IL-4 and IL-25 mRNA in the small intestine, suggesting that once the parasite has been eradicated (see Fig 1), regulatory mechanisms come into play to dampen a mucus/goblet cell response. Interleukin-22 has been implicated in the barrier function of the gut, especially the secretion of anti-microbial factors and mucin [34], and while this can be a direct effect, the diminutation of IL-4 or IL-13 production in the IL-22-/- mice could contribute to the perturbation of mucin and goblet cell regulation following infection with helminth parasites.

Intestinal mast cell hyperplasia can accompany infection with nematodes [24], but c-Kit immunostaining revealed comparable numbers (and distribution) of mast cells in WT and IL-22-/- mice (S2 Fig). These data suggest a limited, if any, role for mast cells in the current study but an in-depth analysis is required before definitive statements on the role of mast cells (with or without IL-22) in the response to H. diminuta can be made.

IL22-/- mice infected with H. diminuta have delayed but enhanced up-regulation of IL-25, IL-10 and Foxp3

Juxtaposing the facts that the epithelium is a target for IL-22 [1] and epithelium-derived factors are important in shaping the immune response and the outcome of infection [35], the impact of the absence of IL-22 on the mobilization of regulatory immune factors/cells was assessed following infection with H. diminuta. The observation of increased IL-25 mRNA in the jejunum of IL-22-/- mice at 8- and 12-dpi with H. diminuta (Fig 2C) suggested that IL-22 serves as a brake on the synthesis of tissue (i.e. epithelial)-derived cytokines elicited in response to infection with helminth parasites. The increase in IL-25 mRNA in the IL-22-/- mice could be due to increased presence of the parasite and not the IL-22-/- deficiency per se. To test this, WT mice were infected with 5 or 10 H. diminuta, and while the latter did lead to increased spleen cell number and TH2 cytokine output, there were no differences in worm burden or intestinal IL-25 mRNA levels between the two infection paradigms at 8-dpi (S3 Fig). Thus, a higher antigenic load is not responsible for the increased IL-25 response but rather this is attributable to the absence of IL-22.

Focusing on IL-25, murine IEC4 epithelial cells were exposed to a single H. diminuta (scolex and ~2 cm of strobila) ± recombinant IL-22. Levels of IL-25 protein and mRNA expression were determined in supernatant and Trizol-treated cells, respectively. The epithelia spontaneously produced IL-25 that was significantly increased by H. diminuta, and in both cases IL-22 reduced IL-25 production (Fig 4A), correlating with mRNA levels (Fig 4B). To our knowledge this is the first time that IL-22 suppression of IL-25 production in the context of a parasitic helminth infection has been shown, underscoring the role of IL-22 in moulding the host response following infection. In addition, using the reductionist approach of culturing a single H. diminuta scolex with epithelial cell lines, we found epithelia from the non-permissive mouse host produced IL-25, IL-33 and TSLP (mRNA and protein) and that the rat (permissive host) IEC.6 cell line failed to show this alarmin response to the worm [36]. Of note, qPCR revealed a trend towards increased IL-33 and thymic stromal lymphopoietin (TSLP) mRNA expression in the jejunum of H. diminuta infected mice (S4 Fig); others have shown differential regulation of IL-25, IL-33 and TSLP following infection with helminth parasites [35].

thumbnail
Fig 4. Interleukin 22 inhibits epithelial IL-25 production.

Monolayers (~3 cm2) of the murine small intestinal IEC4 epithelial cell line were exposed to a single adult H. diminuta (Hd: scolex and 2 cm of strobila) ± recombinant IL-22 (5 ng/ml), and IL-25 measured in the culture medium by ELISA 24 and 48 hr later (Panel A). After collecting supernatants adherent cells were treated with Trizol, total mRNA obtained and IL-25 mRNA expression determined by qPCR (Panel B) (data are mean ± SEM; n = 6; * and #, p<0.05 compared to non-infected control (ctrl) cells and Hd-infected cells, respectively).

https://doi.org/10.1371/journal.ppat.1005481.g004

In addition to its role as a TH2-polarizing cytokine, IL-25 inhibition of trinitrobenzene sulphonic acid (TNBS)-induced colitis in mice may involve alternatively activated macrophages (AAMs) [37], and markers of AAMs are increased in the gut of H. diminuta-infected mice [38]. Extrapolating from this, the increased IL-25 production from epithelia exposed to H. diminuta and the highly significant increase in IL-25 mRNA in the parasitised gut of IL-22-/- could result in increased mobilization of immunoregulatory cells and suppression of concomitant disease in the infected mice.

IL-10 synthesis follows infection with H. diminuta infection and is an important anti-inflammatory cytokine in mice and humans [23]. The increased levels of IL-10, Foxp3 and markers of AAMs (i.e. arginase-1 and Fizz1) mRNA found in the small intestine of H. diminuta-infected Balb/c mice [25], suggests expansion of innate and adaptive regulatory cells. These data were confirmed and extended here and, moreover, gut levels of IL-10 mRNA (Fig 2C) and stimulated IL-10 from splenocytes (Fig 5A) and MLN cells (Fig 5B) were significantly increased at 8- and 12-dpi in IL-22-/- mice compared to WT mice. Macrophages can be an important source of IL-10 in response to helminth and microbial antigens [39, 40]. However, macrophages differentiated from the bone-marrow of IL-22-/- mice had a normal capacity to produce IL-10 in response to H. diminuta antigen or LPS (S5 Fig); thus, we speculate that the increased IL-10 observed in MLN and splenocytes at the later time-points of infection in IL-22-/- mice is from T cells, or potentially B cells [23, 41].

thumbnail
Fig 5. H. diminuta–infected IL-22-/- display delayed and/or prolonged expression of immunoregulatory factors.

Wild-type (WT) and IL-22-/- mice were infected with 5 H. diminuta and at time-points thereafter splenocytes (A) and mesenteric lymph node cells (MLN) (B) were excised and stimulated with conA (5 μg/ml) for 48 hr and IL-10 measured. (C) Shows increases in Foxp3 mRNA in the mid-jejunum of WT and IL-22-/- post-infection. (D) Flow cytometry revealed increased numbers of CD4+Foxp3+ splenocytes 8 days post-infection (dpi) in both WT and IL-22-/- mice (E, representative dot plots) (data are mean ± SEM; n = 6–9; * and # p<0.05 compared to strain control and time-matched WT, respectively).

https://doi.org/10.1371/journal.ppat.1005481.g005

We have shown a variable increase in Foxp3 mRNA in the small intestine of H. diminuta-infected Balb/c mice [25]. Despite the likelihood of IL-22-Foxp3 cross-regulation [42] little is known of the putative interaction of these two factors following infection with helminth parasites. Increased Foxp3 mRNA was observed in the small intestine of IL-22-/- H. diminuta-infected mice compared to WT animals (Fig 5C), supporting the notion that IL-22 serves as a brake on immunoregulatory cell mobilization; however, immunoblotting with extracts of small intestine failed to show a consistent increase in Foxp3+ cells, which may reflect sensitivity of this assay as compared to qPCR (S6 Fig). Moreover, while CD4+Foxp3+ splenocytes were increased following infection (8-dpi) there were no differences between WT and IL-22-/- mice. (Fig 5D and 5E). The reason for the discrepancy between small intestine and spleen is unclear but it underscores the complexity of immunoregulation and the need to precisely define events in both time and space as they relate to the host response to infection. In addition, expression of Foxp3 does not unequivocally identify a cell with immunosuppressive capacity [43], and so IL-10 may be more important than Foxp3 in immunoregulation in this helminth-rodent model system [23].

Although the interaction of IL-25 and Foxp3 expression was not pursued, the association is noteworthy, given data showing lower numbers of Tregs in IL-25-/- mice [44], increases in antigen-specific IL-22+ T cells concomitant with fewer Foxp3+ T cells in an individual with ulcerative colitis infected with T. trichura [22], and that NOD mice treated with IL-25 have increased numbers of Tregs [45]. Thus, one can speculate that the increase in IL-25 in IL-22-/- mice could mediate the increase in Foxp3 and hence Tregs. The role of IL-22 in controlling the mobilization and activity of immunoregulatory cells is not well understood and in addition to considering Tregs, the putative impact of IL-22 on B cells should not be overlooked: for example, successful treatment of tuberculosis correlated with, but was not functionally linked, to increased IL-22 production and a reduced frequency of putative regulatory CD5+CD1d+ B cells [46].

IL-22 participates in DNBS-induced colitis and restricts the anti-colitic effect associated with infection with H. diminuta

Based on the changes observed in IL-22-/- mice following infection with H. diminuta, we examined the impact of lack of IL-22 in (a) the outcome of DNBS-induced colitis and (b) the ability of infection with H. diminuta to reduce the severity of DNBS-induced colitis. IL-22-/- mice consistently developed less severe DNBS-induced colitis compared to WT mice, in all indicies measured: weight loss, colon length and macroscopic appearance, MPO activity (indicative largely of neutrophil infiltration) and the cumulative disease activity score (DAS) (Fig 6). This effect was compounded following infection with H. diminuta: infected IL-22-/- mice treated with DNBS showed minimal signs of disease and were often indistinguishable from control, non-DNBS-treated mice (Fig 6). These findings are in accordance with the reduced mobilization of IFNγ and neutrophils observed in T. gondii-infected IL-22-/- compared to WT mice [47] ((DNBS-induced colitis is considered a TH1-dominated disease and hence the balance of TH1 and TH2 immunity is important in disease severity [48]).

thumbnail
Fig 6. IL-22-/- mice experience less DNBS-induced colitis and enhanced anti-colitic effects following infection with H. diminuta.

Wild-type (WT, gray bars) and IL-22-/- (black bars) mice were infected with 5 H. diminuta (Hd) cysticercoids and 8 days later were given DNBS (5 mg, ir). Mice were necropsied 72 hr post-DNBS and weight loss (A) and degree of colon shortening (B) were assessed for each group. In panel (C), representative colon images from experimental groups are shown. MPO activity in the distal colon was determined and presented as granulocyte marker (D), and overall disease activity scores (DAS) are shown in (E). Data are mean ± SEM; n = 9; * and # p<0.05 compared to strain control and time-matched WT, respectively.

https://doi.org/10.1371/journal.ppat.1005481.g006

Corroborating these macroscopic measures of disease activity, histological analyses revealed that IL-22-/- mice had less DNBS-induced histopathology compared to WT mice, and only very minor damage was observed in the colon of H. diminuta-infected IL-22-/- mice (Fig 7A and 7B). Mitogen stimulation of splenocytes from WT or IL-22-/- mice infected with H. diminuta revealed increased IL-10 production compared to uninfected mice, both naïve and DNBS-treated. Cells from infected DNBS-treated IL-22-/- mice produced, on average, more IL-10 than WT mice, but this did not reach statistical significance (p = 0.2) (Fig 7C). In contrast, splenic production of IL-17, while increased by DNBS, was not significantly different between WT and IL-22-/- mice ± infection with H. diminuta (Fig 7D). Juxtaposing these data with those from H. diminuta-infected naïve IL-22-/- (Figs 25), it is likely that the increase in IL-10, IL-25 and putative regulatory T cells (i.e. increased jejunal Foxp3 mRNA) enhances the anti-colitic effect of infection with H. diminuta in mice lacking IL-22.

thumbnail
Fig 7. Mice lacking IL-22 show reduced DNBS-induced histopathology and increased splenic IL-10.

Wild-type (WT, gray bars) and IL-22-/- (black bars) mice were infected with 5 H. diminuta (Hd) and 8 days later were given DNBS (5 mg, ir). Mice were necropsied 72 hr post-DNBS and histopathology assessed on H&E stained sections (representative images shown in A) and scored on a 12-point scale in a blinded fashion (B). Spleen cells were stimulated in vitro for 48 hr with conA (5 μg/ml) and levels of IL-10 (C) and IL-17 (D) determined by ELISA (data are mean ± SEM; n = 6; * and # p<0.05 compared to strain control and time-matched WT, respectively; M, external muscle layers, L, lumen of colon, scale bar = 100 μm).

https://doi.org/10.1371/journal.ppat.1005481.g007

It has been reported that IL-22 protects female Balb/c mice from TNBS (3 mg, 5 days)-induced colitis [49] (yet others found no increase in IL-22 mRNA in TNBS-treated animals [50]). Given the structural similarity of DNBS and TNBS how can these disparate roles of IL-22 be reconciled? Differences in the sex of mice, the duration of the disease and the natural microbiota of the mice could, at least in part, underlie the opposing findings of the two studies. Also, the protective effect of IL-22 in TNBS-colitis was based on administration of a neutralizing antibody and not genetic knockout of the IL-22 gene, raising the possibility of non-IL-22 effects of the antibody. Again, the point arises that the beneficial versus detrimental impact of manipulating IL-22 as a therapy will be contextual.

IL-22-/- mice have increased susceptibility to dextran sodium sulfate (DSS)-induced colitis [4] and hence the findings in the DNBS model were somewhat surprising. We confirmed that the IL-22-/- mice used here had heightened responsiveness to DSS (S7 Fig). The increased severity of DSS-induced colitis in IL-22-/- mice has been linked to a pro-colitiogenic microbiota [4]. To address this, a published protocol [51] was used to blend the microbiotas between WT and IL-22-/- mice prior to DNBS treatment. The severity of colitis in IL-22-/- mice with their natural microbiota and those who acquired microbiota from WT mice was not different, and both had significantly less disease than WT mice (S8 Fig). In contrast, all of the WT mice who acquired microbiota from IL-22-/- mice presented with severe DNBS-induced colitis, with a marked increase in the size of the cecum: these mice were the sickest of all the experimental groups (S8 Fig). Thus, IL-22-/- mice may harbour a microbial pathobiont that is not important to DNBS-induced colitis in these mice but exaggerates disease in WT mice, somewhat analysis to the transmissibility of susceptibility to DSS by the microbiota from IL-22-/- mice [4]. Assessing the possibility that IL-22-/- could be deficient in anti-microbial peptides, qPCR revealed that this was not the case. In line with findings reported in intestinal bacterial infection increases in mRNA for β-defensin 1, 2 and 3 was similar in WT and knock-out mice following infection with H. diminuta (S9 Fig). Interestingly, unlike infection with C. rodentium that increased RegIIIβ and RegIIIγ in a IL-22-dependent manner [14], infection with H. diminuta evoked only a transient increase in RegIIIβ but not RegIIIγ mRNA (S9 Fig). Thus, the contribution of IL-22 to DNBS-induced colitis is not likely due to different microbiota rather it is a consequence of altered immunoregulation in the absence of IL-22.

The fact that IL-22-/- mice experience less DNBS-induced and greater DSS-induced colitis highlights important differences in disease pathogenesis. Up-regulation of IL-22 mRNA has been found in DSS- but not in TNBS-induced colitis [50]. In the gut, T cells, γδ T cells and ILC3s are major sources of IL-22 [52]. More recently neutrophils have been cited as a source of IL-22 [53]. However, the extent to which each cell is activated in colitis and by which stimuli (i.e. cytokines vs. pattern-associated microbial patterns) is not fully understood. Consequently additional efforts are required to unravel the role of IL-22 in a variety of model systems and in the context of varying microbiotas if extrinsic manipulation of IL-22 levels is to be considered a treatment for enteric disease.

The situation is complicated further by the recent demonstration that IL-25-/- mice are protected from DSS-induced colitis [54] (anti-IL-25 neutralizing antibodies can inhibit oxazolone-induced colitis [55]). Thus, application of anti-IL-22 or anti-IL-25 antibodies to manipulate human disease would need to proceed with caution and be preceded by precise work-up of the immunological basis of the disease in the patient to be treated.

In vivo immunoneutralization of IL-25 in IL-22-/- mice reverses the reduced susceptibility to DNBS-induced colitis

The role of IL-25 has been assessed in TH2-mediated airways diseases as an early TH2-promoting factor [5658]. In the context of TH1-mediated pathologies, IL-25 has been shown to suppress IL-17 and IFNγ production in infectious [26] and autoimmune diseases (e.g experimental autoimmune encephalitis (EAE) [59] and diabetes [45]). Interleukin-25 has been found to inhibit the release of IL-1β, IL-12(p40) and TNFα from LPS-activated human CD14+ monocytes [60] which could in part explain its’ suppression of TH1-driven immunopathologies.

Having found increased IL-25 expression in H. diminuta-infected IL-22-/- mice and that these mice were highly resistant to DNBS-induced colitis, a causal relationship between these two observations was tested via administration of IL-25 neutralizing antibodies [61]. First, the role of IL-25 during DNBS-colitis in IL-22-/- mice in the absence of H. diminuta infection was addressed. Consistent with the previous data, IL-22-/- mice displayed less severe DNBS-induced colitis compared to WT mice (Fig 8). However, IL-22-/- mice treated with DNBS and anti-IL-25 blocking antibodies had a severity of colitis that was macroscopically (Fig 8A–8C) and microscopically (Fig 8D) indistinguishable from WT mice that received DNBS only. Thus, in the absence of infection with H. diminuta (an IL-25 trigger), IL-22 represses IL-25 during inflammatory responses induced by DNBS and when IL-25 is blocked the resistant phenotype observed in IL-22-/- mice is negated.

thumbnail
Fig 8. Resistance to DNBS colitis in IL-22-/- mice requires IL-25.

Wild-type (WT; grey bars) and IL-22-/- (black bars) mice received 5 mg of DNBS intrarectally to induce colitis, with some IL-22-/- also receiving anti-IL-25 blocking antibody (100 μg/mouse, ip., given 10 minutes before DNBS delivery). Severity of colitis was assessed 72 hr post-DNBS by (A, B) measurement of colon length, (C) disease activity score and (D) histological damage score (data are mean ± SEM; n = 6–8; * p<0.05 compared to appropriate strain-matched control mice and # p<0.05 compared to animals given anti-IL-25 blocking antibody).

https://doi.org/10.1371/journal.ppat.1005481.g008

In vivo immunoneutralizing of IL-25 in DNBS+H. diminuta-infected IL-22-/- mice resulted in a severity of colitis that was similar to DNBS-only treated mice, indicating a requirement for IL-25 in the anti-colitic effect evoked following infection with this helminth (Fig 9). These findings complement other studies in which IL-25 has been shown to down-regulate inflammatory gut disease: for example colitis induced in mice by bacterial peptidoglycan, TNBS, oxazolone or DSS [37, 62, 63]. Going forward it will be intriguing to test helminth therapy with/without IL-22 in chronic models of colitis and those driven by adaptive immunity such as the naïve T cell transfer model [4].

thumbnail
Fig 9. Interleukin-25 is a major player in the anti-colitic effect in H. diminuta infected IL-22-/- mice.

Interleukin-22-/- mice were orally infected with 5 cysticercoids of H. diminuta and 8 days later received a single ip. injection of anti-IL-25 antibody (100 μg) concomitantly with delivery of DNBS (5 mg, ir.). Necropsy was performed 72 hr later and severity of colitis determined by (A) colon length, (B) macroscopic disease activity score, (C) histopathology scores, and conA stimulated splenocytes production of (D) IL-4 and (E) IL-10 (data are mean ± SEM; n = 6–8 from 2 independent experiments, except panel E where n = 3–5; * and δ p<0.05 compared to appropriate strain-matched control mice and wild-type (WT) DNBS, respectively) and # p<0.05 compared to animals infected and given anti-IL-25 blocking antibody.

https://doi.org/10.1371/journal.ppat.1005481.g009

Assessment of the role of IL-22 in immunity and inflammation reveals that the impact of this cytokine is highly contextual, with convincing evidence in favour of anti- and pro-inflammatory roles [4, 5, 7, 64]. While many of the functions of IL-22 in the gut promote protective anti-microbial responses, a pathogenic role for IL-22 has been described following infection with T. gondii [19] and Helicobacter pylori [65]. Less is known of the role of IL-22 in the host response to infection with helminth parasites. Increases in local IL-22 or IL-22+ cells have been described in response to gastrointestinal helminths [21, 22], yet the function of IL-22 was inferred not tested. The notable exception being the demonstration of impaired expulsion of nematodes in IL-22-/- mice that aligned with reduced goblet cell hyperplasia [20]. The role of IL-22, if any, in regulating the response to cestode parasites has not hitherto been examined. Production of IL-22 can be evoked by IL-9, IL-23 and microbial stimuli [3] and while IL-25 suppression of IL-22 has been shown [66], less is known of the reciprocal interaction. We have found that increases in IL-25 mRNA in the parasitized intestine and IL-25 synthesis by enteric epithelia exposed to H. diminuta are suppressed by IL-22. This is, to our knowledge, the first time IL-22 has been directly implicated in the control of helminth-evoked IL-25, and complements earlier work showing that IL-22 inhibited IL-25 production by cytokine-treated murine airways epithelia [67].

Using the H. diminuta-mouse model system, data have been obtained that support the following conclusions: (1) absence of IL-22 reduces the early TH2 response to infection with helminth parasites, suggesting an important initial role for innate immunity against metazoan parasites; (2) IL-22 is an endogenous brake on helminth-provoked TH2 immunity, and in its’ absence there is heightened/prolonged local (i.e. gut) and systemic TH2 and immunoregulatory events (e.g. IL-10), likely driven in large part by the increase in IL-25; and, (3) by limiting the synthesis of IL-25, IL-22 participates in the pathogenesis of DNBS-induced colitis and restricts the H. diminuta-suppression of colitis (Fig 10). Helminth therapy has been presented as a novel approach to auto-inflammatory disease [68] and we speculate that precise knowledge of the immunological basis of the disease would be important in selecting patients for helminth therapy.

thumbnail
Fig 10. Proposed model of immune response upon H. diminuta infection in mice lacking IL-22.

The presence of H. diminuta provokes an increase in TH2 and regulatory populations both locally (small intestine) and systemically (MLN and spleen), which ultimately leads to worm expulsion in a non-permissive host (mouse). This regulatory network is responsible for blocking DNBS-colitis. Herein, we identified a heightened regulatory circuit in H. diminuta infected mice lacking IL-22.

https://doi.org/10.1371/journal.ppat.1005481.g010

Methods

Mice, parasites and infection

Interleukin-22 deficient mice (IL-22-/-: C57BL/6 background) were bred at the University of Calgary (pairs kindly provided by Dr. M. Kelly (Univ. of Calgary)). Mice were housed in a 12:12 hr light:dark cycle with free access to food and water and 8–9 weeks old male IL-22-/- and age-matched C57BL/6 control mice (Charles River, QB, Canada) were used throughout this study. As defined in the experiments, mice received 5 infective H. diminuta cysticercoids in 100 μl of sterile 0.9% NaCl by oral gavage and 8 days later colitis was induced [23]. In one experiment doses of 5 and 10 H. diminuta cysticercoids were compared.

Ethics statement

All experiments were conducted following the regulations specified by the Canadian Guidelines for Animal Welfare and were approved by the University of Calgary Health Science Animal Care Committee (HSCCC) with the protocol number AC13-0015.

Worm recovery

At time-points post-infection, the small intestine was excised and flushed with 2 ml of 4°C PBS. The intestine was opened longitudinally and examined along with the flushed contents for H. diminuta.

Induction of experimental colitis and evaluation

Colitis was induced by intrarectal (ir.) installition of 5 mg/mouse of DNBS (MP Biomedicals Ohio, USA) in 100 μl of 50% ethanol 3 cm into the colon. Weight was recorded daily for 3 days, the mice humanely necropsied and a macroscopic disease activity score on a 5 point scale based on weight loss, colon shortening, stool consistency and general appearance determined as previously [23]. A portion of mid-colon was excised, formalin fixed, paraffin embedded and 5 μm sections were collected on coded slided, stained with hematoxylin and eosin and a histopathology score determined on a 12-point scale [23]. The most distal 1 cm of colon was snap frozen in liquid nitrogen for myeloperoxidase (MPO) determination as measure of granulocyte, mainly neutrophil, infiltrate. MPO activity was determined by a kinetic assay in which H2O2 catabolism is measured, and 1 unit of MPO activity is the amount of enzyme required to degrade 1 μM of H2O2/min [23].

In other experiments, a 5 day exposure to 2.5% wt./vol. DSS (MW: 30,000–50,000; MP, Biomedicals, OH, USA) was used to induce colitis. Mice were transferred to regular tap water on day 5, and 3 days later were assessed for disease severity as described above.

Goblet cell and mast cell staining

Formalin-fixed, paraffin-embedded mouse mid-small intestine was sectioned (5 μm), sections collected on coded slides and stained with periodic-acid Schiff’s stain to identify goblet cells [31]. Cells were counted on a per villus-crypt unit (VCU) basis, as defined by an intact, rounded villus tip and an even layer or enterocytes indicating lack of obligue sectioning.

To identify mast cells, sections were deparaffinized followed by epitope retrieval with 10 mM sodium citrate buffer pH 6.0. After washing sections were incubated in PE anti-mouse CD117 (c-Kit) antibody (BioLegend, CA, USA) (1:500) in blocking solution at 4°C overnight. Subsequently sections were washed in PBS, incubated in DAPI (0.1 μg/mL, 10 min. at room temperatura) and after a final PBS wash, slides were mounted using ProLong Gold (Cell Signaling Technology) and examined with a Nikon 80i microscope and DXM1200C camera. Images were captured using NIS-Elements software (Nikon), and representative images were processed in Adobe Photoshop (Version 8.0).

Measurement of systemic immune response

At indicated times the spleen and mesenteric lymph nodes (MLN) were asceptically removed from WT and IL-22-/- H. diminuta-infected mice, cell suspensions generated and red blood cells lysed in ammonium chloride buffer [23]. Cells were adjusted to 3x106 /ml in RPMI 1640 medium supplemented with 10% FBS, 0.1 mM (Gibco, USA). Cells were activated by treatment with concanavalin A (5 μg/ml) and 48 hr later supernatants were collected and stored (-80°C) for cytokine measurements by ELISA.

ELISA sandwich

Interleukin (IL)-4, IL-5, IL-10, IL-13, IL-17, IL-25 and IFNγ were measured by ELISA using paired antibodies and following the manufactures’ instructions (R&D Systems Inc., Minneapolis, USA). All samples were measured in duplicate and assays had dectection limits that ranged from 2–9 pg/ml.

Flow cytometry

Spleens were aseptically excised and cell suspensions generated as above. Thereafter, 1x106 splenocytes were incubated with TrueStainX (anti-CD16/32) for 10 min at 4°C and then stained. Cells were stained for 30 min with conjugated APC-CD4 (Biolegend, San Diego, CA USA). After incubation with APC-CD4 antibody cells were washed in flow buffer (PBS, 1%FBS and 0.1% NaN3) and intracellular staining for Foxp3 was performed following manufacturer’s protocol. Briefly, after surface staining cells were washed with flow cytometry buffer, then fixed and permeabilized with Foxp3 Fix/Perm and Foxp3 Perm buffers respectively. A final incubation with Foxp3-AlexaFluor 488 (Biolegend, San Diego, CA) was conducted for 30 min at room temperature in the dark. Data were acquired in a Attune cytometer and analyzed with Attune V.6.1 software (R&D systems).

qPCR in intestine

Small intestine was excised from non-infected and H. diminuta-infected WT and IL-22-/- mice, flushed with 4°C PBS, and the 3 cm portion of mid-intestine was cut in three pieces, placed in 1ml of TRizol Reagent (Invitrogen, California, USA) and homogenized for 60 seconds (Polytron MR2100, Kinematica AG, Switzerland). The RNA was extracted with chloroform/ethanol as previously [25] and 1 μg of RNA was used as the template for cDNA generation with the iScript DNA synthesis kit (Bio-Rad, USA). Conditions for the PCR were denaturation 95°C for 2 min, 40 amplifying cycles of 95°C 15 sec, 55°C 15 sec, 68°C 20 sec and final temperature 4°C; primer sequences are presented in S10 Fig.

Immunoblotting for Foxp3

At indicated times after H. diminuta infection ~1cm of jejunum was excised and homogenized in RIPA buffer (50 mM Tris-HCl, 150 mM NaCl, 1% NP-40 0.5% sodium deoxycholate and 0.1% SDS) supplemented with protease inhibitor cocktail (Promega, Madison Wisconsin USA). Protein concentration was determined by the Bradford assay (Bio-Rad Laboratories Mississauga ON, Canada). Samples were normalized to 10 μg protein/μl and run by SDS-page (4% stacking, 8% separating) and transferred to a nitrocellulose membrane. Membranes were blocked for 1 hr at room temperature in 5% skim milk in 0.1% TBS-tween buffer and then incubated overnight with purified anti-Foxp3, 3 μg/ml (Biolegend, California, USA). After washing, membranes were incubated with appropriate secondary antibody for 1 hr at room temperature and developed by exposing to western lightning plus enhanced chemiluminiscence solution (PerkinElmer, Woodbridge ON, Canada) for 1 min and using an automatic film developer.

Exposure of IEC4 cells to live H. diminuta

The mouse small intestinal epithelial cell line, IEC4, was maintained by serial passage in DMEM medium supplemented with HEPES (1%), L-glutamine (10%), Pen/Strep (1%) and FBS (5%) (all from Gibco, USA). One-million IEC4 cells were seeded in 6-well plates and cultured for 48 hr. Scolices and 2 cm of strobila of H. diminuta retrieved from the small intestine of rats or IL-4 receptor-α-/- mice (fail to expel H. diminuta) were exposed to a cocktail of antibiotics (Gentamicin solution, Sigma, St. Louis, Mo, USA) for 2 hr. A single worm was added to IEC4 monolayers ± recombinant IL-22 (5 ng/ml; Biolegend, CA, USA), and supernatants collected for measurement of IL-25 and then total RNA extracted.

In vivo IL-25 neutralization

To determine the role of IL-25, IL-22-/- were treated with a single ip. injection of 100 μg of an anti-IL-25 blocking antibody (clone 35B, Biolegend, CA, USA) ~10 min prior to DNBS ir. delivery and the severity of colon inflammation was assessed 72 hr later (as above [23]).

Transfer of colonic bacteria

Following a protocol to transfer colonic microbiota between mice [51,69], WT and IL-22-/- mice were transferred to cages with fresh bedding and 24 hr later mice were swapped into the opposing strains cage without a bedding change for 24 hr (coprophagy allows blending of the microbiota between the two strains). This cycle of swapping mice between cages was continued for 2 weeks. On day one of the procedure all mice were treated with kanamycin (40 mg/kg), gentamicin (3.5 mg/kg), colistin (4.2 mg/kg), metronidazole (21.5 mg/kg) for 3 days in their drinking water followed by an ip. injection of vancomycin (4.5 mg/kg). On day 15 mice were anesthetized and given DNBS (5 mg/mouse) intrarectally and colitis severity was assessed 72 hr later.

Bone marrow derived macrophage development and in vitro stimulation

Bone marrow was flushed from the long bones of the legs of WT and IL-22-/- mice via a sterile 27 gauge needle, the red blood cells lysed and the cells were incubated in RPMI 1640 medium (Gibco, USA) supplemented with 20% FBS, HEPES, Glutamax and antibiotic (Penicillin-streptomycin Sigma, St. Louis, Mo, USA) for 7 days in presence of 20 ng/ml murine M-CSF. On days 2 and 4 cells were treated with fresh medium containing macrophage-colony stimulating factor (M-CSF). At day 7, mature macrophages were harvested and seeded at 2.5x105 in 24-well plates in above-mentioned medium and incubated with PBS-soluble crude H. diminuta antigen (HdAg: 100 μg/ml [70]) for 24 hr. As additional control, macrophages were also stimulated with LPS (10–1000 ng/ml (Sigma, St. Louis, MO, USA)). Supernatants were collected and assayed for TNFα by ELISA.

Statistical analysis

Data are presented as mean ± the standard error of the mean (SEM) and statistical differences were determined by one-way ANOVA followed by post-hoc analysis with Student’s t test or Kneuman’s Keuls test and p<0.05 accepted as a statistically significant difference (Graph Pad prism V5 software, La Jolla, CA, USA).

Supporting Information

S1 Fig. Levels of TH1 cytokine IFN γ decrease during H. diminuta infection and remain unaltered in absence of IL-22.

At 4, 8 and 12 days post infection spleen from infected experimental groups were collected, RBCs depleted and cell suspensions generated. Cell suspensions were incubated for 48 hr in presence of conA (5 μg/ml) and supernatants collected. Levels of IFN γ were determined by ELISA as described in methods. Data shown are mean ± SEM from independent experiments where * p<0.05 as compared to strain-matched control and # p<0.05 compared to WT time-matched group (n = 7).

https://doi.org/10.1371/journal.ppat.1005481.s001

(TIF)

S2 Fig. Mast cells numbers are not significantly different in H. diminuta-infected wild-type (WT) or IL-22-/- mice.

Mice were necropsied at the days post-infection (dpi) indicated, ~1cm of mid-jejunum was collected, fixed, paraffin embedded and immuno-staining performed with anti-cKit antibody (mast cell marker), as per the manufacturer’s instructions, and DAPI staining used to identify nuclei. Random fields of view were chosen based on DAPI staining and observed in a blinded fashion (images are representative of n = 3–4 mice; original mag. = x200).

https://doi.org/10.1371/journal.ppat.1005481.s002

(TIF)

S3 Fig. High parasite burden results in increased cellular response but comparable IL-25 mRNA expression in small intestine.

Wild-type mice received 5 or 10 cysticercoids of H. diminuta and on necropsy 8 days later (A) there was no difference in worm expulsion, while (B-D) the number of splenocytes and concanavalin-induced IL-4 and IL-10 production was significantly increased. (E) However, analysis of mid-jejunum segments extracted in Trizol by qPCR revealed no differences in IL-25 mRNA expression. Lines represent mean ± SEM; n = 5; * p<0.05 as compared to animals infected with 5 cysticercoids.

https://doi.org/10.1371/journal.ppat.1005481.s003

(TIF)

S4 Fig. Tissue-derived cytokines IL-33 and TSLP are not modified due to IL-22 absence.

Total mRNA was extracted from small intestine on indicated times after H. diminuta infection from both WT and IL-22-/- mice and (A) IL-33 and (B) TSLP transcripts were measured and normalized against the housekeeping gene 18s. Data shown are mean ± SEM from 2 independent experiments (n- = 6).

https://doi.org/10.1371/journal.ppat.1005481.s004

(TIF)

S5 Fig. Macrophages from IL-22-/- mice do not over-produce IL-10 in response to H. diminuta antigens.

Bone marrow precursors from WT and IL-22-/- mice were differentiated into macrophages for 7 days as described in methods. Upon additional 24 hr of stimulation, supernatans were collected and levels of IL-10 in response to H. diminuta crude antigens (A) and LPS (B) were determined by ELISA. Data shown are from 2 independent experiments with similar results (n = 6).

https://doi.org/10.1371/journal.ppat.1005481.s005

(TIF)

S6 Fig. Protein levels of Foxp3+ showed no increase in IL-22-/- mice compared to WT counterparts.

At indicated times post-infection small intestine tissue from both WT and IL-22-/- mice was homogenized in RIPA buffer and total protein extraction was conducted as indicated in methods and Foxp3 protein levels were determined. Beta-actin was used as loading control. Image is representative of 2 experiments with similar results.

https://doi.org/10.1371/journal.ppat.1005481.s006

(TIF)

S7 Fig. IL-22-/- mice are more susceptible to DSS-induced colitis than WT mice.

Wild-type (WT) and IL-22-/- mice were exposed to 2.5% (wt./vol.) dextran sodium sulfate (DSS) for 5 days followed by 3 days of normal drinking water and on necropsy IL-22-/- mice displayed increased disease severity as assessed by (A) weight loss, (B) colon length, and (C) disease activity scores (DAS) (data are mean ± SEM; n = 5; * and #, p<0.05 compared to appropriate strain control (ctrl) and WT DSS mice, respectively).

https://doi.org/10.1371/journal.ppat.1005481.s007

(TIF)

S8 Fig. Resistance to DNBS-induced colitis observed in IL-22-/- mice is not due to their microbiota.

The microbiotas were blended (Mix. Mic.) between wild-type (WT; gray bars) and IL-22-/- mice (black bars) by cross-cage exchange and exploiting the coprophagic behavior of mice, followed by DNBS (5 mg, ir, 72 hr) treatment. The mixed or blended microbiota in IL-22-/- mice did not affect their susceptibility to DNBS, with both groups having less severe colitis than WT mice assessed by colon shortening (A) and overall macroscopic score (B). Representative colon images in (C) show a reduced severity in IL-22-/- mice regardless of having acquired microbiota from WT mice. In contrast, WT mice receiving microbiota from IL-22-/- mice had the most severe colitis. Also, analysis of blind-scored H&E colon sections (D), confirmed less histopathological damage in absence of IL-22. Data are mean ± SEM; n = 5; * and #, p<0.05 compared to the appropriate strain matched control naïve mice and WT DNBS mice, respectively; arrow indicates enlarged caecum.

https://doi.org/10.1371/journal.ppat.1005481.s008

(TIF)

S9 Fig. IL-22-/- mice did not show impaired defensin and RegIII peptides expression during H. diminuta infection.

Mid-jejunum tissue was homogenized in Trizol at the indicated time points and mRNA extracted as described in methods. Gene expression of Defensins 1–3 (A) and Reg III beta and gamma peptides was determined by using the specific primers quoted in S10 Fig Data are mean ± SEM from 2 independent experiments (n = 6), p< 0.0.5 as compared to expression found in wild-type (WT) animals.

https://doi.org/10.1371/journal.ppat.1005481.s009

(TIF)

S10 Fig. Primer sequences used to determine gene expression of highly relevant players of intestinal immunity.

Sequences were syntethized in Univ. of Calgary DNA core facilities or when indicated (i.e. β defensin 2 and β defensin 3) sequences were obtained as ready-to-use primer assay from Qiagen. Muc; mucin, Reg; Regeneration islet-derived protein.

https://doi.org/10.1371/journal.ppat.1005481.s010

(TIF)

Acknowledgments

We gratefully acknowledge the core facilities provided by the Snyder Institute for Chronic Disease and the Flow Cytometry suite at the Univ. Calgary.

Author Contributions

Conceived and designed the experiments: DMM JLR. Performed the experiments: JLR MRF FL GL NLM CEM AW. Analyzed the data: DMM JLR. Wrote the paper: DMM JLR.

References

  1. 1. Leung JM, Loke P. A role for IL-22 in the relationship between intestinal helminths, gut microbiota and mucosal immunity. Int J Parasitol. 2013;43(3–4):253–7. pmid:23178750
  2. 2. Sabat R, Ouyang W, Wolk K. Therapeutic opportunities of the IL-22-IL-22R1 system. Nature reviews Drug discovery. 2014;13(1):21–38. Epub 2014/01/01. pmid:24378801
  3. 3. Dudakov JA, Hanash AM, van den Brink MR. Interleukin-22: immunobiology and pathology. Annual review of immunology. 2015;33:747–85. pmid:25706098
  4. 4. Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy AJ, Stevens S, Flavell RA. Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease. Immunity. 2008;29(6):947–57. Epub 2008/12/23. pmid:19100701
  5. 5. Mielke LA, Jones SA, Raverdeau M, Higgs R, Stefanska A, Groom JR, et al. Retinoic acid expression associates with enhanced IL-22 production by γδ T cells and innate lymphoid cells and attenuation of intestinal inflammation. The Journal of experimental medicine. 2013;210(6):1117–24. pmid:23690441
  6. 6. Sugimoto K, Ogawa A, Mizoguchi E, Shimomura Y, Andoh A, Bhan AK, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. The Journal of clinical investigation. 2008;118(2):534–44. Epub 2008/01/04. pmid:18172556
  7. 7. Eken A, Singh AK, Treuting PM, Oukka M. IL-23R+ innate lymphoid cells induce colitis via interleukin-22-dependent mechanism. Mucosal immunology. 2014;7(1):143–54. pmid:23715173
  8. 8. Leung JM, Davenport M, Wolff MJ, Wiens KE, Abidi WM, Poles MA, et al. IL-22-producing CD4+ cells are depleted in actively inflamed colitis tissue. Mucosal immunology. 2014;7(1):124–33. pmid:23695510
  9. 9. Longman RS, Diehl GE, Victorio DA, Huh JR, Galan C, Miraldi ER, et al. CX(3)CR1(+) mononuclear phagocytes support colitis-associated innate lymphoid cell production of IL-22. The Journal of experimental medicine. 2014;211(8):1571–83. pmid:25024136
  10. 10. Radaeva S, Sun R, Pan HN, Hong F, Gao B. Interleukin 22 (IL-22) plays a protective role in T cell-mediated murine hepatitis: IL-22 is a survival factor for hepatocytes via STAT3 activation. Hepatology. 2004;39(5):1332–42. Epub 2004/05/04. pmid:15122762
  11. 11. Zhang Y, Cobleigh MA, Lian JQ, Huang CX, Booth CJ, Bai XF, et al. A proinflammatory role for interleukin-22 in the immune response to hepatitis B virus. Gastroenterology. 2011;141(5):1897–906. Epub 2011/06/29. pmid:21708106
  12. 12. Justa S, Zhou X, Sarkar S. Endogenous IL-22 plays a dual role in arthritis: regulation of established arthritis via IFN-γ responses. PloS one. 2014;9(3):e93279. Epub 2014/03/29. pmid:24676270
  13. 13. Pineda MA, Rodgers DT, Al-Riyami L, Harnett W, Harnett MM. ES-62 protects against collagen-induced arthritis by resetting interleukin-22 toward resolution of inflammation in the joints. Arthritis & rheumatology. 2014;66(6):1492–503.
  14. 14. Zheng Y, Valdez PA, Danilenko DM, Hu Y, Sa SM, Gong Q, et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nature medicine. 2008;14(3):282–9. pmid:18264109
  15. 15. Wilson MS, Feng CG, Barber DL, Yarovinsky F, Cheever AW, Sher A, et al. Redundant and pathogenic roles for IL-22 in mycobacterial, protozoan, and helminth infections. Journal of immunology. 2010;184(8):4378–90. Epub 2010/03/12.
  16. 16. Behnsen J, Jellbauer S, Wong CP, Edwards RA, George MD, Ouyang W, et al. The cytokine IL-22 promotes pathogen colonization by suppressing related commensal bacteria. Immunity. 2014;40(2):262–73 pmid:24508234
  17. 17. De Luca A, Zelante T, D'Angelo C, Zagarella S, Fallarino F, Spreca A, et al. IL-22 defines a novel immune pathway of antifungal resistance. Mucosal immunology. 2010;3(4):361–73. pmid:20445503
  18. 18. Kagami S, Rizzo HL, Kurtz SE, Miller LS, Blauvelt A. IL-23 and IL-17A, but not IL-12 and IL-22, are required for optimal skin host defense against Candida albicans. Journal of immunology. 2010;185(9):5453–62.
  19. 19. Munoz M, Heimesaat MM, Danker K, Struck D, Lohmann U, Plickert R, et al. Interleukin (IL)-23 mediates Toxoplasma gondii-induced immunopathology in the gut via matrixmetalloproteinase-2 and IL-22 but independent of IL-17. The Journal of experimental medicine. 2009;206(13):3047–59. pmid:19995958
  20. 20. Turner JE, Stockinger B, Helmby H. IL-22 mediates goblet cell hyperplasia and worm expulsion in intestinal helminth infection. PLoS pathogens. 2013;9(10):e1003698. pmid:24130494
  21. 21. Gaze S, McSorley HJ, Daveson J, Jones D, Bethony JM, Oliveira LM, et al. Characterising the mucosal and systemic immune responses to experimental human hookworm infection. PLoS pathogens. 2012;8(2):e1002520. pmid:22346753
  22. 22. Broadhurst MJ, Leung JM, Kashyap V, McCune JM, Mahadevan U, McKerrow JH, et al. IL-22+ CD4+ T cells are associated with therapeutic Trichuris trichiura infection in an ulcerative colitis patient. Science translational medicine. 2010;2(60):60ra88. Epub 2010/12/03. pmid:21123809
  23. 23. Hunter MM, Wang A, Hirota CL, McKay DM. Neutralizing anti-IL-10 antibody blocks the protective effect of tapeworm infection in a murine model of chemically induced colitis. Journal of immunology. 2005;174(11):7368–75. Epub 2005/05/21.
  24. 24. Maizels RM, Hewitson JP, Smith KA. Susceptibility and immunity to helminth parasites. Current opinion in immunology. 2012;24(4):459–66. pmid:22795966
  25. 25. Persaud R, Wang A, Reardon C, McKay DM. Characterization of the immuno-regulatory response to the tapeworm Hymenolepis diminuta in the non-permissive mouse host. International journal for parasitology. 2007;37(3–4):393–403. Epub 2006/11/10. pmid:17092505
  26. 26. Owyang AM, Zaph C, Wilson EH, Guild KJ, McClanahan T, Miller HR, et al. Interleukin 25 regulates type 2 cytokine-dependent immunity and limits chronic inflammation in the gastrointestinal tract. The Journal of experimental medicine. 2006;203(4):843–9. pmid:16606667
  27. 27. Angkasekwinai P, Srimanote P, Wang YH, Pootong A, Sakolvaree Y, Pattanapanyasat K, et al. Interleukin-25 (IL-25) promotes efficient protective immunity against Trichinella spiralis infection by enhancing the antigen-specific IL-9 response. Infection & Immunity 2013;81(10):3731–41.
  28. 28. Zaiss MM, Maslowski KM, Mosconi I, Guenat N, Marsland BJ, Harris NL. IL-1β suppresses innate IL-25 and IL-33 production and maintains helminth chronicity. PLoS pathogens. 2013;9(8):e1003531. pmid:23935505
  29. 29. Marillier RG, Michels C, Smith EM, Fick LC, Leeto M, Dewals B, et al. IL-4/IL-13 independent goblet cell hyperplasia in experimental helminth infections. BMC immunology. 2008;9:11. pmid:18373844
  30. 30. Onah DN, Nawa Y. Mucosal immunity against parasitic gastrointestinal nematodes. The Korean journal of parasitology. 2000;38(4):209–36. pmid:11138315
  31. 31. McKay DM, Halton DW, McCaigue MD, Johnston CF, Fairweather I, Shaw C. Hymenolepis diminuta: intestinal goblet cell response to infection in male C57 mice. Experimental Parasitology 1990;71(1):9–20. pmid:2354717
  32. 32. Johansson ME, Sjovall H, Hansson GC. The gastrointestinal mucus system in health and disease. Nature reviews Gastroenterology & hepatology. 2013;10(6):352–61.
  33. 33. Webb RA, Hoque T, Dimas S. Expulsion of the gastrointestinal cestode, Hymenolepis diminuta by tolerant rats: evidence for mediation by a Th2 type immune enhanced goblet cell hyperplasia, increased mucin production and secretion. Parasite immunology. 2007;29(1):11–21. pmid:17187651
  34. 34. Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. IL-22 increases the innate immunity of tissues. Immunity. 2004;21(2):241–54. pmid:15308104
  35. 35. Hepworth MR, Danilowicz-Luebert E, Rausch S, Metz M, Klotz C, Maurer M, et al. Mast cells orchestrate type 2 immunity to helminths through regulation of tissue-derived cytokines. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(17):6644–9. pmid:22493240
  36. 36. Lopes F, Reyes JL, Wang A, Leung G, McKay DM. Enteric epithelial cells support growth of Hymenolepis diminuta in vitro and trigger TH2-promoting events in a species-specific manner. International journal for parasitology. 2015;45(11):691–6. pmid:26151388
  37. 37. Rizzo A, Monteleone I, Fina D, Stolfi C, Caruso R, Fantini MC, et al. Inhibition of colitis by IL-25 associates with induction of alternatively activated macrophages. Inflammatory bowel diseases. 2012;18(3):449–59. pmid:21688353
  38. 38. Hunter MM, Wang A, Parhar KS, Johnston MJ, Van Rooijen N, Beck PL, et al. In vitro-derived alternatively activated macrophages reduce colonic inflammation in mice. Gastroenterology. 2010;138(4):1395–405. pmid:20044996
  39. 39. Bode JG, Ehlting C, Haussinger D. The macrophage response towards LPS and its control through the p38(MAPK)-STAT3 axis. Cellular signalling. 2012;24(6):1185–94. pmid:22330073
  40. 40. Wi HJ, Jin Y, Choi MH, Hong ST, Bae YM. Predominance of IL-10 and TGF-β production from the mouse macrophage cell line, RAW264.7, in response to crude antigens from Clonorchis sinensis. Cytokine. 2012;59(2):237–44. pmid:22579699
  41. 41. Ziegler T, Rausch S, Steinfelder S, Klotz C, Hepworth MR, Kuhl AA, et al. A novel regulatory macrophage induced by a helminth molecule instructs IL-10 in CD4+ T cells and protects against mucosal inflammation. Journal of immunology. 2015;194(4):1555–64.
  42. 42. Mathian A, Parizot C, Dorgham K, Trad S, Arnaud L, Larsen M, et al. Activated and resting regulatory T cell exhaustion concurs with high levels of interleukin-22 expression in systemic sclerosis lesions. Annals of the rheumatic diseases. 2012;71(7):1227–34. pmid:22696687
  43. 43. Morikawa H, Ohkura N, Vandenbon A, Itoh M, Nagao-Sato S, Kawaji H, et al. Differential roles of epigenetic changes and Foxp3 expression in regulatory T cell-specific transcriptional regulation. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(14):5289–94. pmid:24706905
  44. 44. Donaldson DS, Bradford BM, Artis D, Mabbott NA. Reciprocal regulation of lymphoid tissue development in the large intestine by IL-25 and IL-23. Mucosal immunology. 2014.
  45. 45. Emamaullee JA, Davis J, Merani S, Toso C, Elliott JF, Thiesen A, et al. Inhibition of Th17 cells regulates autoimmune diabetes in NOD mice. Diabetes. 2009;58(6):1302–11. pmid:19289457
  46. 46. Zhang M, Zeng G, Yang Q, Zhang J, Zhu X, Chen Q, et al. Anti-tuberculosis treatment enhances the production of IL-22 through reducing the frequencies of regulatory B cell. Tuberculosis. 2014;94(3):238–44. pmid:24566282
  47. 47. Munoz M, Eidenschenk C, Ota N, Wong K, Lohmann U, Kuhl AA, et al. Interleukin-22 Induces Interleukin-18 Expression from Epithelial Cells during Intestinal Infection & Immunity. 2015;42(2):321–31. pmid:25680273
  48. 48. Khan WI, Motomura Y, Wang H, El-Sharkawy RT, Verdu EF, Verma-Gandhu M, et al. Critical role of MCP-1 in the pathogenesis of experimental colitis in the context of immune and enterochromaffin cells. American journal of physiology gastrointestinal and liver physiology. 2006;291(5):G803–11. pmid:16728728
  49. 49. Monteleone I, Rizzo A, Sarra M, Sica G, Sileri P, Biancone L, et al. Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract. Gastroenterology. 2011;141(1):237–48, 48 e1. pmid:21600206
  50. 50. te Velde AA, de Kort F, Sterrenburg E, Pronk I, ten Kate FJ, Hommes DW, et al. Comparative analysis of colonic gene expression of three experimental colitis models mimicking inflammatory bowel disease. Inflammatory bowel diseases. 2007;13(3):325–30. pmid:17206675
  51. 51. Lopes F, Wang A, Smyth D, Reyes JL, Doering A, Schenck LP, et al. The Src kinase Fyn is protective in acute chemical-induced colitis and promotes recovery from disease. Journal of Leukocyte Biology. 2015;97(5)1089–99.
  52. 52. Wolk K, Sabat R. Interleukin-22: a novel T- and NK-cell derived cytokine that regulates the biology of tissue cells. Cytokine & growth factor reviews. 2006;17(5):367–80.
  53. 53. Zindl CL, Lai JF, Lee YK, Maynard CL, Harbour SN, Ouyang W, et al. IL-22-producing neutrophils contribute to antimicrobial defense and restitution of colonic epithelial integrity during colitis. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(31):12768–73. pmid:23781104
  54. 54. Reynolds JM, Lee YH, Shi Y, Wang X, Angkasekwinai P, Nallaparaju KC, et al. Interleukin-17B antagonizes interleukin-25-mediated mucosal inflammation. Immunity. 2015;42(4):692–703. pmid:25888259
  55. 55. Camelo A, Barlow JL, Drynan LF, Neill DR, Ballantyne SJ, Wong SH, et al. Blocking IL-25 signalling protects against gut inflammation in a type-2 model of colitis by suppressing nuocyte and NKT derived IL-13. Jouranl of Gastroenterology 2012;47(11):1198–211.
  56. 56. Tamachi T, Maezawa Y, Ikeda K, Kagami S, Hatano M, Seto Y, et al. IL-25 enhances allergic airway inflammation by amplifying a TH2 cell-dependent pathway in mice. The journal of allergy and clinical immunology. 2006;118(3):606–14. pmid:16950278
  57. 57. Fort MM, Cheung J, Yen D, Li J, Zurawski SM, Lo S, et al. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity. 2001;15(6):985–95. pmid:11754819
  58. 58. Ballantyne SJ, Barlow JL, Jolin HE, Nath P, Williams AS, Chung KF, et al. Blocking IL-25 prevents airway hyperresponsiveness in allergic asthma. The journal of allergy and clinical immunology. 2007;120(6):1324–31. pmid:17889290
  59. 59. Kleinschek MA, Owyang AM, Joyce-Shaikh B, Langrish CL, Chen Y, Gorman DM, et al. IL-25 regulates Th17 function in autoimmune inflammation. The journal of experimental medicine. 2007;204(1):161–70. pmid:17200411
  60. 60. Caruso R, Stolfi C, Sarra M, Rizzo A, Fantini MC, Pallone F, et al. Inhibition of monocyte-derived inflammatory cytokines by IL-25 occurs via p38 Map kinase-dependent induction of Socs-3. Blood. 2009;113(15):3512–9. pmid:19129540
  61. 61. Angkasekwinai P, Park H, Wang YH, Wang YH, Chang SH, Corry DB, et al. Interleukin 25 promotes the initiation of proallergic type 2 responses. The journal of experimental medicine. 2007;204(7):1509–17. pmid:17562814
  62. 62. Caruso R, Sarra M, Stolfi C, Rizzo A, Fina D, Fantini MC, et al. Interleukin-25 inhibits interleukin-12 production and Th1 cell-driven inflammation in the gut. Gastroenterology. 2009;136(7):2270–9. pmid:19505427
  63. 63. McHenga SS, Wang D, Li C, Shan F, Lu C. Inhibitory effect of recombinant IL-25 on the development of dextran sulfate sodium-induced experimental colitis in mice. Cellular & molecular immunology. 2008;5(6):425–31.
  64. 64. Kamanaka M, Huber S, Zenewicz LA, Gagliani N, Rathinam C, O'Connor W Jr., et al. Memory/effector (CD45RB(lo)) CD4 T cells are controlled directly by IL-10 and cause IL-22-dependent intestinal pathology. The Journal of experimental medicine. 2011;208(5):1027–40. pmid:21518800
  65. 65. Zhuang Y, Cheng P, Liu XF, Peng LS, Li BS, Wang TT, et al. A pro-inflammatory role for Th22 cells in Helicobacter pylori-associated gastritis. Gut. 2014.
  66. 66. Sawa S, Lochner M, Satoh-Takayama N, Dulauroy S, Berard M, Kleinschek M, et al. RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nature immunology. 2011;12(4):320–6. pmid:21336274
  67. 67. Takahashi K, Hirose K, Kawashima S, Niwa Y, Wakashin H, Iwata A, et al. IL-22 attenuates IL-25 production by lung epithelial cells and inhibits antigen-induced eosinophilic airway inflammation. The journal of allergy and clinical immunology. 2011;128(5):1067–76 e1-6. pmid:21794904
  68. 68. Fleming JO, Weinstock JV. Clinical trials of helminth therapy in autoimmune diseases: rationale and findings. Parasite immunology. 2015;37(1):277–92.
  69. 69. Chen X, Katchar K, Goldsmith JD, Nanthakumar N, Cheknis A, Gerding DN, et al. A mouse model of Clostridium difficile-associated disease. Gastroenterology. 2008;135(6):1984–92. pmid:18848941
  70. 70. Johnston MJ, Wang A, Catarino ME, Ball L, Phan VC, MacDonald JA, et al. Extracts of the rat tapeworm, Hymenolepis diminuta, suppress macrophage activation in vitro and alleviate chemically induced colitis in mice. Infect Immun. 2010;78(3):1364–75. pmid:20028812