Skip to main content
Erschienen in: Journal of Gastroenterology 4/2018

Open Access 26.10.2017 | Review

Clinical importance of IL-22 cascade in IBD

verfasst von: Atsushi Mizoguchi, Arisa Yano, Hidetomo Himuro, Yui Ezaki, Takayuki Sadanaga, Emiko Mizoguchi

Erschienen in: Journal of Gastroenterology | Ausgabe 4/2018

Abstract

IL-22 is a relatively new cytokine that is characterized by several unique biological properties. In the intestines, the effect of IL-22 is restricted mainly to non-lymphoid cells such as epithelial cells. Interestingly, the expression pattern and major cellular source of IL-22 have distinct difference between large and small intestines. IL-22 possesses an ability to constitutively activate STAT3 for promoting epithelial cell regeneration and reinforcing mucosal barrier integrity through stimulating the expression of anti-bacterial peptide and mucins. Of note, IL-22 is characterized as a two-faced cytokine that can play not only protective but also deleterious roles in the intestinal inflammation depending on the cytokine environment such as the expression levels of IL-23, T-bet, and IL-22 binding protein. Most importantly, clinical relevance of IL-22 to inflammatory bowel disease has been well highlighted. Mucosal healing, which represents the current therapeutic goal for IBD, can be induced by IL-22. Indeed, indigo naturalis, which can activate IL-22 pathway through Ahr, has been shown in a clinical trial to exhibit a strong therapeutic effect on ulcerative colitis. Despite the beneficial effect of IL-22, continuous activation of the IL-22 pathway increases the risk of colitis-associated cancer, particularly in patients with an extended history of IBD. This review article discusses how IL-22 regulates colitis, how beneficial versus deleterious effects of IL-22 is determined, and why IL-22 represents a promising target for IBD therapy.
Abkürzungen
Ahr
Aryl hydrocarbon receptor
CAC
Colitis-associated cancer
DC
Dendritic cells
DSS
Dextran sulfate sodium
IBD
Inflammatory bowel disease
IL-22BP
IL-22 binding protein
ILC
Innate lymphoid cells
Muc
Mucin
Reg
Regenerating gene
RORγt
RAR-related orphan receptor γt
STAT
Signal transducers and activator of transcription
TCR
T cell receptor
UC
Ulcerative colitis

Introduction

Interleukin (IL)-22 is a relatively new cytokine that was discovered in 2000 in both humans and mice [1, 2]. IL-22 was identified in humans as a new ligand of IL-10R2 that can activate STATs 1, 3, and 5 [1] and simultaneously identified in mice as an IL-10-related T cell-inducible factor that was detected in thymic lymphomas stimulated with IL-9 [2]. Rapid progress in the IL-22 biology has since been made in basic and clinical science, both of which have demonstrated its unique properties [36]. For example, expression of IL-22 receptor (IL-22R) is restricted to non-hematopoietic cells such as intestinal and respiratory epithelial cells, keratinocytes, and hepatocytes, allowing IL-22 to specifically target innate immune responses without directly affecting adaptive immune cells [7]. Of note, the function of IL-22 has been figuratively called “a sheep in wolf’s clothing” [8] or “a two-headed cytokine” [4] due to its dual abilities to not only exacerbate but also improve inflammatory responses. In addition, IL-22 exhibits unique expression pattern particular in the intestine. IL-22 is constitutively expressed in the small intestine to maintain the epithelial integrity that provides the first line of defense against enteric microorganisms for preserving appropriate host–microbial interactions [9, 10]. In contrast, IL-22 represents an inducible cytokine in the large intestine where IL-22 expression is hardly detectable under healthy state and can be induced under inflammatory conditions such as inflammatory bowel disease (IBD) [1113]. The clinical relevance of IL-22 to IBD has been well highlighted by the fact that the majority of IL-22-associated molecules are encoded by IBD susceptibility genes [3]. In addition, IL-22 signaling may provide a clue to solve a long-standing epidemiologic mystery in IBD regarding a negative association of cigarette smoking with the development of ulcerative colitis (UC).

Distinct IL-22 expression pattern in small versus large intestine

There are distinct expression patterns of IL-22 between small and large intestines [3]. IL-22 is constitutively expressed in the small intestine of humans and mice to maintain epithelial barrier integrity against enteric microorganisms that play an important role in the pathogenesis of IBD [9, 10, 14]. In contrast, IL-22 expression, which is rarely detectable in the large intestine (colon) of humans and mice under healthy condition [1113, 1517], is induced by a wide variety of inflammatory conditions ranging from IBD to infection [1113, 1517]. However, the expression level of inducible IL-22 may differ depending on the types of inflammation. For example, the expression level of IL-22 in the inflamed colon is higher in Crohn’s disease (CD) patients as compared to UC patients [11, 12, 15]. Similarly, much higher expression level of IL-22 has been reported in the inflamed colon of a mouse model of CD (CD45RB model) as compared to a mouse model of UC (TCRα KO mice) [12]. CD is characterized by dominant Th1/Th17 responses, whereas UC is associated with enhanced Th2 response [18]. In addition, atopic dermatitis, which exhibits a dominant Th2 response, is associated with lower levels of IL-22 expressions as compared to psoriasis characterized by enhanced Th1/Th17 responses [19]. Therefore, it is possible that IL-22 expression, which is elicited by inflammatory insults, is further conditioned by the local cytokine environments (e.g., Th1 versus Th2).

Cellular sources of IL-22 in the intestine

IL-22 can be expressed by many cell types, including Th17 CD4+ T cells that express both IL-17 and IL-22, Th22 CD4+ T cells that express IL-22 without producing IL-17, CD8+ T cells, TCRγδ T cells, dendritic cells (DCs), neutrophils, NK cells, and group3 innate lymphoid cell (ILC) population including ILC3 and lymphoid tissue inducer (LTi) cells [46, 20]. The major cellular source of IL-22 in the small intestine is RAR-related orphan receptor (ROR)γt-dependent ILC3 that can develop in the absence of enteric bacteria as indicated by the development of ILC3 in germ-free mice [14, 21]. In Toxoplasma gondii infection, the major source of IL-22 in the small intestine shifts from ILC3 to CD4+ T cells [22].
In human colon, different types of cells may be responsible for the inflammation-induced production of IL-22 depending on the type of inflammation and presumably on the disease stages. The ability of NK cells and Th17 T cells to produce IL-22 was initially reported in the inflamed colon of UC patients [16], and subsequent studies have suggested that IL-22-producing NKp46+ NKp44 NK cells more increase in the inflamed colon of CD patients as compared to that of UC patients [23]. A recent study has found an additional cell type that can be responsible for the production of IL-22 in IBD. CD177+ neutrophils produce IL-22 for improving colitis and this neutrophil subset increases in the peripheral blood and inflamed intestine of UC and CD patients [20]. In CD patients, CD4+ T cells characterized by the expression of natural killer group 2 member D (NKG2D) have been reported to produce IL-22 [24]. Group 3 ILC population is further classified into ILC3 that produces IL-22 and expresses CD56 and LTi cells that produce both IL-17 and IL-22 without expression of CD56 [25]. An increases of both IL-22-producing CD56+ ILC3 and IL-17-producing CD56 LTi cells are observed in the inflamed colon of CD patients as compared to UC patients [26]. Alternatively, ILC3 has recently been shown to possess an additional function to serve as immune suppressive antigen presenting cells capable of eliminating enteric bacteria-specific colitogenic CD4+ T cells through an MHC class II-dependent manner [27]. In the inflamed colon of pediatric CD patients, the total frequency of ILC3 cells is not altered, but their expression level of MHC class II is reduced [28].
Similar to IBD patients, cellular sources of IL-22 vary in the inflamed colon of mouse IBD models depending on the fundamental disease mechanisms. In a mouse model of CD (CD45RB transfer model), both NK cells and Th17 cells are primarily responsible for the production of IL-22 in the inflamed colon [13]. Alternatively, dendritic cells (DCs) [17], neutrophils [29], and/or TCRγδ T cells [30] represent the major source of IL-22 in an acute colonic injury model, which is induced by oral administration of dextran sulfate sodium (DSS). DCs have also been suggested as a major source of IL-22 in C. rodentium- infectious colitis model [31]. In contrast, a study suggested in the C. rodentium infectious model that RORγt+ LTi cells, but not DCs, produce IL-22 in the inflamed colon [32]. In addition, IL-23-dependent LTi cells, which are characterized by the localization within the intraepithelial compartment of colon, have been proposed to represent a dominant source of colonic IL-22 in the context of C. rodentium infection [33]. Taken together, these findings indicate that IL-22 can be produced by many cell types depending on the tissue involved and on the healthy versus disease states.

Epithelial cell-specific STAT3 activation by IL-22

STAT3 is one of the major signaling molecules involved in the pathogenesis of IBD. However, STAT3 possesses dual abilities to not only improve but also exacerbate intestinal inflammation. Activation of STAT3 in adaptive immune cells such as CD4+ T cells contributes for the development of colitis in a mouse model of CD [34]. On the other hand, activation of STAT3 in innate immune cells such as macrophages contributes to the suppression of colitis as indicated by the spontaneous development of colitis in a conditional knockout mouse strain in which the stat3 gene is specifically deleted in macrophages/neutrophils [35]. In addition, absence of STAT3 in epithelial cells makes mice more susceptible to DSS-induced acute colonic injury [17]. Therefore, to pay careful attention would be needed when STAT3 is considered as a target for the treatment of IBD.
IL-22 activates (phosphorylates) STAT3, to a lesser extent STAT1, and in certain cells STAT5 [36, 37]. This STAT3 activation is mediated by tyrosine kinase 2 (Tyk2) [38]. In addition, IL-22 has been reported to activate Erk1/2, JNK, and p38 MAP in some, not all, epithelial cell lines. A high dose of IL-22 activates Erk1/2, JNK, and p38 MAP kinase in a rat hepatoma cell line [36, 37] and human colonic cancer cell line HT29 [15]. In contrast, inability of IL-22 to activate Erk/1/2 has been shown in a human hepatoma cell line HepG2 [39] and primary epithelial cells isolated freshly from human and mouse colons [12]. IL-22 binds to a heterodimeric receptor composed of IL-22R1 and IL-10R2 [46]. IL-10R2 is ubiquitously expressed by majority of cell types, while the expression of IL-22R1 is restricted to non-hematopoietic cells such as epithelial cells, hepatocytes, and keratinocytes [46]. This expression pattern of IL-22R1 allows IL-22 to activate STAT3 specifically in intestinal epithelial cells [7]. Indeed, epithelial STAT3 activation induced by intestinal damage has been demonstrated to depend more on IL-22 rather than IL-6, a well-known activator of STAT3 [17]. This strong activation of STAT3 by IL-22 may be explained by the ability of IL-22R1 to constitutively, not transiently, activate STAT3 through constitutive interaction of its C-terminal tail with the coiled-coil domain of STAT3 [40].

Production of anti-bacterial peptides by IL-22

Dysregulated host–microbial interactions are closely involved in the pathogenesis of IBD. Epithelial cells provide the first barrier against enteric microorganisms, and the ability of IL-22 to reinforce the epithelial barrier integrity has been well documented [3]. IL-22 stimulates the production of different kinds of anti-bacterial peptides such as S100A7, S100A8, S100A9 β-defensin 2, and regenerating gene (Reg) IIIγ and RegIIIβ [17, 31, 4143]. One of well-studied anti-bacterial peptides in the field of IL-22 may be RegIII family. Bactericidal ability of RegIIIγ against Gram-positive bacteria [44] was first discovered in 2006, and a subsequent study using RegIIIα identified that the bactericidal effect is mediated by the formation of a hexameric membrane-permeabilizing oligomeric pore on Gram-positive bacteria and the bactericidal effect is inhibited by Gram-negative bacteria-derived lipopolysaccharide [45]. Alternatively, a protective role of RegIIIγ and RegIIIβ against Gram-negative bacteria C. rodentium has also been proposed [31]. There still remains an issue under debate regarding whether RegIII expression is mediated directly by IL-22-dependent STAT3 activation or not. Several investigators suggested the induction of RegIII γ and β expressions through IL-22-dependent STAT3 activation [17], whereas others suggested the NFκB1-dependent RegIIIγ expression through IL-23 or MyD88 [46, 47]. A recent study has brought an attractive new concept that RegIIIβ represents the initiation, rather than downstream, molecule in IL-22 pathway. RegIIIβ is, at first, produced by epithelial cells through IL-23 receptor signaling, and the epithelial-derived RegIIIβ then recruits IL-22-producing neutrophils that contribute for the improvement of colitis [48].
Other IL-22-associated anti-bacterial peptides that have clinical relevance may be S100A8 and S100A9. S100A8 and S100A9 form a heterodimer complex termed calprotectin, and fecal calprotectin has been widely used as a biomarker for IBD [49]. Although calprotectin is produced mainly by neutrophils, inflammation can induce the ectopic expression of calprotectin in colonic epithelial cells by IL-22 [42]. In addition, expressions of S100A8 and S100A9 are mediated by STAT3 activation [50]. Therefore, it is possible that the fecal calprotectin may reflect not only neutrophil-derived calprotectin but also ectopically expressed calprotectin in epithelial cells by inflammation-induced IL-22.

Reinforcement of mucus barrier integrity by IL-22

Mucus, which covers the intestinal surface, also plays a major role in preserving epithelial barrier integrity [51]. The mucus layer in the colon is composed of outer and inner stratum; the outer is a loose gel layer where it is unattached to the epithelial surface and provides a place for colonized bacteria, whereas the inner stratum is a firm mucus layer that is directly attached to the epithelial surface to prevent bacterial intrusion [52]. Mucin (Muc) 1, which is a membrane-bound mucin, represents one of the major components in the intestinal mucus [51]. A protective role of Muc1 in mouse models of UC and CD has been demonstrated [53]. The ability of IL-22 to promote the production of functional Muc1 through activation of STAT3 has been demonstrated using human colonic cancer cell lines (T84 and HT29) and primary colonic epithelial cells from mice [12, 54]. Consistent with these findings, STAT3 has been demonstrated to bind within the promoter region of the muc1 gene [55]. These findings from in vitro and mouse studies are further supported by data from IBD patients. An early onset IBD patient who has no functional IL-10R2 (a receptor of IL-22) due to polymorphism within il10rb gene has been reported to lack Muc1 expression [56]. Importantly, Muc1 is a heavily glycosylated protein with over 80% (per total molecular weight) of carbohydrates that are required to form a viscous firm barrier. Therefore, not only the expression of Muc1 but also its complete glycosylation are required to preserve epithelial barrier integrity. Indeed, overexpression of hypoglycosylated Muc1 makes breaches in the mucus barrier [57] and exacerbates colitis [58]. Of note, IL-22 has been demonstrated to promote glycosylation [43, 59]. Therefore, it is possible that the dual abilities to stimulate not only Muc1 expression but also glycosylation allow IL-22 to produce completely firm inner mucus layer to prevent bacterial invasion.

Mucosal healing by IL-22

IL-22-dependent activation of STAT3 enhances the transcription of anti-apoptotic and pro-proliferative genes such as birc5, pla2g5, smo, myc, mcl1and regIα [16, 60]. The expression of deleted in malignant brain tumor 1 (DMBT1), which may play a role in epithelial cell differentiation, is also induced by IL-22 [61]). These abilities allow IL-22 to promote mucosal healing by stimulating epithelial cell regeneration with goblet cell restitution [12, 17]. In addition, recent studies have demonstrated that the epithelial cell regeneration is initiated by activation of intestinal stem cells through IL-22-dependent activation of STAT3 but not STAT1 [62, 63].
Consistent with these basic observations, the ability of IL-22 to promote mucosal healing has been well documented in vivo by using different experimental approaches. Mucosal healing after epithelial damage induced by DSS was impaired in IL-22-deficient mice [17], IL-23R-deficient mice that lack IL-22 expressions [47], and WT mice treated with neutralizing anti-IL-22 Abs [12]. In addition, a gene therapy to enhance colonic IL-22 expression [12] and an antibody-based targeted delivery of IL-22 to inflamed area [64] promoted the mucosal healing. The mucosal healing was also enhanced by the treatment with Ficz, which is a ligand of Ahr capable of inducing the expression of IL-22 [65]. Since the induction and maintenance of mucosal healing is the major therapeutic goal in the management of IBD [66], the ability to promote mucosal healing clearly highlights the relevance of IL-22 for IBD therapy.

Newly identified functions of IL-22

Attractive new functions of IL-22 to induce diarrhea for the clearance of enteric pathogens, to dampen adaptive immune responses against enteric bacterial antigens, and to improve endoplasmic reticulum (ER) stress have recently been demonstrated. IL-22 stimulated the expression of a tight junction molecule Claudin-2, which induces water efflux (diarrhea) for the clearance of enteric pathogens [67]. In addition, IL-22 suppressed the antigen uptake of follicular associated epithelium (FAE) that covers Peyer’s and colonic patches where they provide a place for the induction of adaptive immune responses against enteric antigens [68]. Furthermore, IL-22 was shown to reduce epithelial ER/oxidative stress induced by high-fat diets [69].

Dual roles of IL-22 in intestinal inflammation

As referred to as “a sheep in wolf’s clothing” [8] or “a two-headed cytokine” [4], not only protective but also deleterious roles of IL-22 have been reported in intestinal inflammation models [3]. In infectious models, IL-22-deficient mice are highly susceptible to intestinal inflammation caused by Citrobacter rodentium [14, 31, 33, 70], Salmonella enterica [71], and candidiasis [72]. In contrast, Toxoplasma gondii-induced immunopathology was improved in the absence of IL-22 [22].
Although the involvement of IL-22 in mucosal healing has been reported reproducibly by many groups, the different roles of IL-22 have been demonstrated in chronic colitis models. In a Th2-mediated chronic colitis model (TCRα KO mice) representing UC, IL-22 was shown to play a protective role by reinforcing intestinal mucus barrier function [12]. In a Th1-mediated CD model (CD45RB model) that is induced by the adoptive transfer of naïve CD4+ T cells into a immune-deficient host, both T cell-derived and NK cell-derived IL-22 contributed for the suppression of colitis [13]. Ficz, which is a ligand of Ahr capable of inducing IL-22 expression, was reported to improve the colitis of the CD45RB model as well as acute CD model induced by intrarectal administration of trinitrobenzene sulfonic acid (TNBS) [65]. Alternatively, in a chronic colitis model that is induced by the adoptive transfer of memory CD4+ T cells, a proinflammatory role of memory T cell-derived IL-22 was proposed [73]. Similarly, IL-23R-dependent IL-22 production caused intestinal inflammation in an innate colitis model that was induced in immune-deficient mice by administration of anti-CD40 mAbs [74].

Control of IL-22 functions by IL-23R and T-bet

It’s becoming increasingly apparent that the function of IL-22 is positively and negatively regulated by the complicated interactions with other factors (Fig. 1). One of the nice examples may be psoriasis, in which the pathogenic potential of IL-22 is elicited only in the presence of IL-17 and IFN-γ [75]. Similarly, IL-22 plays a pro-inflammatory role in the presence of IL-17A and conversely plays a protective role in the absence of IL-17A in bleomycin-induced airway inflammation [76]. In an IBD model, IL-22 plays a pathogenic role under a condition where T cells are hyper responsive to IL-23 for their Th17 differentiation due to absence of T-bet (a transcription factor for Th1 differentiation) [77]. Alternatively, early induction of IL-23 and IL-1β, although both are pro-inflammatory cytokines, are required for initiating the IL-22-dependent tissue repair [78]. In addition, IL-23 signaling in epithelial cells licenses the protective effect of IL-22 on intestinal inflammation. IL-23R-mediated signaling stimulates epithelial cells to produce RegIIIβ, which then recruits IL-22-producing neutrophils capable of improving colitis [48]. Interestingly, an autocrine regulation of IL-22 pathway has also been proposed recently. The RegIIIβ produced by epithelial cells, in turn, inhibits their STAT3 activation in response to IL-22 [79].

An endogenous inhibitor of IL-22

Activation of IL-22 is tightly and directly regulated by an endogenous inhibitor. IL-22 binds to not only a heterodimeric receptor composed of IL-10R2 and IL-22R1 but also a soluble class II cytokine receptor designated IL-22Ra2 (also called IL-22 binding protein, IL-22BP, or CRF2-10). In comparison with IL-22R1, IL-22BP has 20–1000 fold higher affinity to IL-22 [8082]. Therefore, IL-22BP can strongly inhibit the biological activity of IL-22 in vitro and in vivo by competing with IL-22R1/IL-22 interaction [12, 8082]. IL-22BP is highly expressed in placenta, spleen, skin, and lung, and to a lesser extent in large and small intestine of humans [80]. In mouse intestine, DCs [83], CD4+ T cells [84], and epithelial cells [85] have been proposed as a producer of IL-22BP. The IL-22BP is highly expressed in the normal colon and significantly reduced during acute inflammation induced by DSS [12, 83, 86]. The inflammation-induced reduction of IL-22BP expression is caused by a formation of inflammasomes that function as a sensor of stress to activate IL-1 and IL-18 [83] or by the maturation of DCs, suggesting immature DCs as a source of IL-22BP [86]. In contrast to mice, rather increased expression of intestinal IL-22BP was observed in patients with IBD, and CD4+ T cells [84] and eosinophils [87] have been proposed as the cellular source of IL-22BP. However, more caution may be necessary to evaluate the expression of functional IL-22BP, because there are three functionally different isoforms of IL-22BP in humans but not mice [88]. Isoform 1, which is composed of six exons, is non-functional due to the lack of secretion. Isoform 2, lacking exon 3, represents a homologue of mouse IL-22BP and possesses a strong inhibitory effect on IL-22 activity. Isoform 3, which lacks exons 3 and 5 and partially exon 6, is most abundantly distributed, but the affinity of isoform 3 to IL-22 is 27-folds lower than that of isoform 2 [88].
Since IL-22BP expression became rarely detectable during the acute inflammatory response induced by DSS [12, 83], deficiency of IL-22BP in mice had no significant alteration on the acute inflammation [83]. Alternatively, supplementation of IL-22BP expression delayed the recovery from this acute inflammation by inhibiting IL-22 activity [12]. In a chronic colitis model, TNF-α stimulated the IL-22BP expression in CD4+ T cells, leading to the suppression of IL-22-dependent mucosal healing [84]. Indeed, anti-TNF-α treatment reduced the expression of IL-22BP in CD4+ T cells of IBD patients [84]. Therefore, this finding may provide a clue to further understand the therapeutic mechanism of anti- TNF-α therapy.

IL-22-associated IBD susceptibility gene network

Several facts have highlighted the clinical significance of IL-22 to IBD. One of the examples is that the majority of IL-22-associated molecules are encoded by IBD susceptibility genes. IL-23 is a heterodimeric cytokine composed of p40 and p19 subunits. The interaction of IL-23 with IL-23R has been demonstrated to not only stimulate IL-22 expression [5, 6], but also positively and negatively control the function of IL-22 [77, 78]. Il23r gene represents one of the major IBD susceptibility genes and the polymorphism was negatively associated with the development of both CD and UC [89, 90]. In addition, p40 polymorphism was also associated with the development of CD [91].
The receptor of IL-22 is composed of IL-10Rβ and IL-22R1 subunits. Positive association of il10rb polymorphisms with CD and UC particularly with early onset of IBD has been well documented [92]. In addition, the il22 gene is located within a UC-risk locus on chromosome 12q15 [93], and il22 polymorphism in UC patients has been reported in the Chinese [94], but not Mexican [95], population. The ligation of IL-22 receptor is well known to induce the activation of STAT3 through JAK2 and TYK2. The stat3, jak2, and tyk2 all represent well-defined susceptibility genes for CD and to lesser extent UC [89, 90]. The STAT3 activation then stimulates the colonic epithelial cells to produce Muc1 and Muc13 for barrier reinforcement [11, 12, 55]. The muc13 gene is an IBD susceptibility gene, and the muc1 gene has been proposed as a candidate susceptible gene of CD [96]. In addition, the ability of IL-22 to directly or indirectly promote the expressions of fut2, sec1, bcl2115, ptpn22, prdm1, xbp1, nupr1, erbp3, efemp2, chac1, pbbp, cxcl5, and sk9a3 has recently been demonstrated by a study using IL-22Ra1-deficient mice [43]. Importantly, all of them have been reported as IBD susceptibility genes. Taken together, these findings highlight the close involvement of IL-22 in the molecular network associated with IBD susceptibility genes.

A clue to resolve an etiological mystery in UC

A negative association of cigarette smoking with the development UC has been well documented [97]. However, the mechanism is a long-standing question, thus considered as one of ten remaining mysteries in IBD [98]. Aryl hydrocarbon receptor (Ahr), which serves as a sensor that can be activated by a large variety of environmental toxins, can induce the production of IL-22 [99]. Indeed, like IL-22, the beneficial effect of Ahr activation on experimental colitis has been demonstrated [65]. Cigarette smoke contains more than 150 toxins, including dioxin such as 2,3,7,8-tetrachlorodibenzo-p-doxin (TCDD) and environmental pollutants such as polycyclic aryl hydrocarbons, of which benzo [α] pyrene (BaP) is a prototype [100]. Of note, TCDD and BaP can activate the Ahr [100], and induction of IL-22 expression by cigarette smoke has recently been demonstrated [101]. These findings may raise an attractive hypothesis that the preventive effect of smoking on UC development is mediated by Ahr-dependent activation of IL-22 pathway.

Therapeutic application of IL-22

Lactobacilli species metabolize tryptophan to indole-3-acetic acid that can activate Ahr for stimulating IL-22 production [102]. Therapeutic benefit of tryptophan on experimental colitis through activation of the IL-22 pathway has also been demonstrated [102, 103]. In addition, the fecal from UC and CD patients was characterized by a low level of tryptophan and indole-3-acetic acid [103].
Xilei-San is a Chinese herbal medicine that has been used for the treatment of UC [104]. Xilei-San contains a high concentration of indigo naturalis, a dye extracted from plants. A recent open-label, prospective pilot study on 20 patients with moderate UC showed that oral administration of indigo naturalis induced mucosal healing in 61% of patients at 8 weeks [105]. Importantly, indigo naturalis is one of the ligands of Ahr, and a recent study in mice demonstrates that the therapeutic effect of indigo naturalis is mediated by Ahr-dependent activation of IL-22 pathway [106]. In addition, the therapeutic potential of other Ahr ligands such as Ficz [65] and ABX464 (the first-in-class anti-HIV drug candidate) [107] against experimental colitis has been reported. These findings clearly highlight the activation of IL-22 as a promising therapeutic measure for IBD, particularly UC. However, more caution may be needed when supplementation of IL-22 is considered for IBD therapy. Chronic inflammation predisposes IBD patients to the risk of colitis-associated cancer that develops through a complex multi-step and multi-factorial processes termed “inflammation-dysplasia-carcinoma sequence” [108]. Hyper activation of STAT3 is one of factors involved in the development of CAC [109], and continuous activation of IL-22 due to lack of IL-22BP has been shown to increase the susceptibility to CAC induced by chronic intestinal inflammation plus a carcinogen AOM [83]. These findings suggest that IL-22/IL-22BP axis critically regulates mucosal healing and tumorigenesis.

Conclusions

IL-22 is characterized by numerous clinical relevance to IBD (Fig. 2), including the ability to induce the mucosal healing that represents the current therapeutic goal of IBD, the close involvement in IBD susceptibility gene networks, and the association with CAC, the potential role in an etiological mystery of UC, and the stimulation of expression of a biomarker for IBD. In addition, the IL-22 pathway seems to mediate the therapeutic effect of indigo naturalis and partially of anti-TNFα therapy, both of which have been used for the treatment of IBD. Although much more extensive works still need to fully dissect out the mechanism that determines the beneficial versus deleterious effect of IL-22 on colitis and that stimulates the tumorigenesis pathway, IL-22 activation would be considered to be a promising therapeutic measures for IBD.

Acknowledgements

We would like to greatly thank Ms. Satomi Uchida for her excellent editorial assistance. This project was supported by NIH R01 DK091247 (AM and EM) and by Grants-in-Aid for Scientific Research (B) 15H04813.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Xie MH, Aggarwal S, Ho WH, et al. Interleukin (IL)-22, a novel human cytokine that signals through the interferon receptor-related proteins CRF2-4 and IL-22R. J Biol Chem. 2000;275:31335–9.PubMedCrossRef Xie MH, Aggarwal S, Ho WH, et al. Interleukin (IL)-22, a novel human cytokine that signals through the interferon receptor-related proteins CRF2-4 and IL-22R. J Biol Chem. 2000;275:31335–9.PubMedCrossRef
2.
Zurück zum Zitat Dumoutier L, Louahed J, Renauld JC. Cloning and characterization of IL-10-related T cell-derived inducible factor (IL-TIF), a novel cytokine structurally related to IL-10 and inducible by IL-9. J Immunol. 2000;164:1814–9.PubMedCrossRef Dumoutier L, Louahed J, Renauld JC. Cloning and characterization of IL-10-related T cell-derived inducible factor (IL-TIF), a novel cytokine structurally related to IL-10 and inducible by IL-9. J Immunol. 2000;164:1814–9.PubMedCrossRef
4.
Zurück zum Zitat Seiderer J, Brand S. IL-22: a two-headed cytokine in IBD? Inflamm Bowel Dis. 2009;15:473–4.PubMedCrossRef Seiderer J, Brand S. IL-22: a two-headed cytokine in IBD? Inflamm Bowel Dis. 2009;15:473–4.PubMedCrossRef
5.
Zurück zum Zitat Ouyang W, Rutz S, Crellin NK, et al. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu Rev Immunol. 2011;29:71–109.PubMedCrossRef Ouyang W, Rutz S, Crellin NK, et al. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu Rev Immunol. 2011;29:71–109.PubMedCrossRef
6.
Zurück zum Zitat Sonnenberg GF, Fouser LA, Artis D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol. 2011;12:383–90.PubMedCrossRef Sonnenberg GF, Fouser LA, Artis D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol. 2011;12:383–90.PubMedCrossRef
7.
Zurück zum Zitat Wolk K, Kunz S, Witte E, et al. IL-22 increases the innate immunity of tissues. Immunity. 2004;21:241–54.PubMedCrossRef Wolk K, Kunz S, Witte E, et al. IL-22 increases the innate immunity of tissues. Immunity. 2004;21:241–54.PubMedCrossRef
8.
Zurück zum Zitat Laurence A, O’Shea JJ, Watford WT. Interleukin-22: a sheep in wolf’s clothing. Nat Med. 2008;14:247–9.PubMedCrossRef Laurence A, O’Shea JJ, Watford WT. Interleukin-22: a sheep in wolf’s clothing. Nat Med. 2008;14:247–9.PubMedCrossRef
9.
Zurück zum Zitat Sanos SL, Bui VL, Mortha A, et al. RORgammat and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat Immunol. 2009;10:83–91.PubMedCrossRef Sanos SL, Bui VL, Mortha A, et al. RORgammat and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat Immunol. 2009;10:83–91.PubMedCrossRef
10.
Zurück zum Zitat Cella M, Fuchs A, Vermi W, et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature. 2009;457:722–5.PubMedCrossRef Cella M, Fuchs A, Vermi W, et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature. 2009;457:722–5.PubMedCrossRef
11.
Zurück zum Zitat Andoh A, Zhang Z, Inatomi O, et al. Interleukin-22, a member of the IL-10 subfamily, induces inflammatory responses in colonic subepithelial myofibroblasts. Gastroenterology. 2005;129:969–84.PubMedCrossRef Andoh A, Zhang Z, Inatomi O, et al. Interleukin-22, a member of the IL-10 subfamily, induces inflammatory responses in colonic subepithelial myofibroblasts. Gastroenterology. 2005;129:969–84.PubMedCrossRef
12.
Zurück zum Zitat Sugimoto K, Ogawa A, Mizoguchi E, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 2008;118:534–44.PubMedPubMedCentral Sugimoto K, Ogawa A, Mizoguchi E, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 2008;118:534–44.PubMedPubMedCentral
13.
Zurück zum Zitat Zenewicz LA, Yancopoulos GD, Valenzuela DM, et al. Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease. Immunity. 2008;29:947–57.PubMedPubMedCentralCrossRef Zenewicz LA, Yancopoulos GD, Valenzuela DM, et al. Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease. Immunity. 2008;29:947–57.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Lee JS, Cella M, McDonald KG, et al. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat Immunol. 2011;13:144–51.PubMedPubMedCentralCrossRef Lee JS, Cella M, McDonald KG, et al. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat Immunol. 2011;13:144–51.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Brand S, Beigel F, Olszak T, et al. IL-22 is increased in active Crohn’s disease and promotes proinflammatory gene expression and intestinal epithelial cell migration. Am J Physiol Gastrointest Liver Physiol. 2006;290:G827–38.PubMedCrossRef Brand S, Beigel F, Olszak T, et al. IL-22 is increased in active Crohn’s disease and promotes proinflammatory gene expression and intestinal epithelial cell migration. Am J Physiol Gastrointest Liver Physiol. 2006;290:G827–38.PubMedCrossRef
16.
Zurück zum Zitat Sekikawa A, Fukui H, Suzuki K, et al. Involvement of the IL-22/REG Ialpha axis in ulcerative colitis. Lab Invest. 2010;90:496–505.PubMedCrossRef Sekikawa A, Fukui H, Suzuki K, et al. Involvement of the IL-22/REG Ialpha axis in ulcerative colitis. Lab Invest. 2010;90:496–505.PubMedCrossRef
17.
Zurück zum Zitat Pickert G, Neufert C, Leppkes M, et al. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J Exp Med. 2009;206:1465–72.PubMedPubMedCentralCrossRef Pickert G, Neufert C, Leppkes M, et al. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J Exp Med. 2009;206:1465–72.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448:427–34.PubMedCrossRef Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448:427–34.PubMedCrossRef
19.
Zurück zum Zitat Guttman-Yassky E, Lowes MA, Fuentes-Duculan J, et al. Low expression of the IL-23/Th17 pathway in atopic dermatitis compared to psoriasis. J Immunol. 2008;181:7420–7.PubMedPubMedCentralCrossRef Guttman-Yassky E, Lowes MA, Fuentes-Duculan J, et al. Low expression of the IL-23/Th17 pathway in atopic dermatitis compared to psoriasis. J Immunol. 2008;181:7420–7.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Zhou G, Yu L, Fang L,et al. CD177+ neutrophils as functionally activated neutrophils negatively regulate IBD. Gut. 2017. PMID:28468761 (In Press) Zhou G, Yu L, Fang L,et al. CD177+ neutrophils as functionally activated neutrophils negatively regulate IBD. Gut. 2017. PMID:28468761 (In Press)
21.
Zurück zum Zitat Sawa S, Cherrier M, Lochner M, et al. Lineage relationship analysis of RORgammat + innate lymphoid cells. Science. 2010;330:665–9.PubMedCrossRef Sawa S, Cherrier M, Lochner M, et al. Lineage relationship analysis of RORgammat + innate lymphoid cells. Science. 2010;330:665–9.PubMedCrossRef
22.
Zurück zum Zitat Muñoz M, Heimesaat MM, Danker K, et al. Interleukin (IL)-23 mediates Toxoplasma gondii-induced immunopathology in the gut via matrixmetalloproteinase-2 and IL-22 but independent of IL-17. J Exp Med. 2009;206:3047–59.PubMedPubMedCentralCrossRef Muñoz M, Heimesaat MM, Danker K, et al. Interleukin (IL)-23 mediates Toxoplasma gondii-induced immunopathology in the gut via matrixmetalloproteinase-2 and IL-22 but independent of IL-17. J Exp Med. 2009;206:3047–59.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Takayama T, Kamada N, Chinen H, et al. Imbalance of NKp44(+)NKp46(−) and NKp44(−)NKp46(+) natural killer cells in the intestinal mucosa of patients with Crohn’s disease. Gastroenterology. 2010;139:882–92.PubMedCrossRef Takayama T, Kamada N, Chinen H, et al. Imbalance of NKp44(+)NKp46(−) and NKp44(−)NKp46(+) natural killer cells in the intestinal mucosa of patients with Crohn’s disease. Gastroenterology. 2010;139:882–92.PubMedCrossRef
24.
Zurück zum Zitat Pariente B, Mocan I, Camus M, et al. Activation of the receptor NKG2D leads to production of Th17 cytokines in CD4+ T cells of patients with Crohn’s disease. Gastroenterology. 2011;141:217–26.PubMedCrossRef Pariente B, Mocan I, Camus M, et al. Activation of the receptor NKG2D leads to production of Th17 cytokines in CD4+ T cells of patients with Crohn’s disease. Gastroenterology. 2011;141:217–26.PubMedCrossRef
25.
Zurück zum Zitat Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells—how did we miss them? Nat Rev Immunol. 2013;13:75–87.PubMedCrossRef Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells—how did we miss them? Nat Rev Immunol. 2013;13:75–87.PubMedCrossRef
26.
Zurück zum Zitat Geremia A, Arancibia-Cárcamo CV, Fleming MP, et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J Exp Med. 2011;208:1127–33.PubMedPubMedCentralCrossRef Geremia A, Arancibia-Cárcamo CV, Fleming MP, et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J Exp Med. 2011;208:1127–33.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Hepworth MR, Monticelli LA, Fung TC, et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature. 2013;498:113–7.PubMedPubMedCentralCrossRef Hepworth MR, Monticelli LA, Fung TC, et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature. 2013;498:113–7.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Hepworth MR, Fung TC, Masur SH, et al. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4+ T cells. Science. 2015;348:1031–5.PubMedPubMedCentralCrossRef Hepworth MR, Fung TC, Masur SH, et al. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4+ T cells. Science. 2015;348:1031–5.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Zindl CL, Lai JF, Lee YK, et al. IL-22-producing neutrophils contribute to antimicrobial defense and restitution of colonic epithelial integrity during colitis. Proc Natl Acad Sci USA. 2013;110:12768–73.PubMedPubMedCentralCrossRef Zindl CL, Lai JF, Lee YK, et al. IL-22-producing neutrophils contribute to antimicrobial defense and restitution of colonic epithelial integrity during colitis. Proc Natl Acad Sci USA. 2013;110:12768–73.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Mielke LA, Jones SA, Raverdeau M, et al. Retinoic acid expression associates with enhanced IL-22 production by γδ T cells and innate lymphoid cells and attenuation of intestinal inflammation. J Exp Med. 2013;210:1117–24.PubMedPubMedCentralCrossRef Mielke LA, Jones SA, Raverdeau M, et al. Retinoic acid expression associates with enhanced IL-22 production by γδ T cells and innate lymphoid cells and attenuation of intestinal inflammation. J Exp Med. 2013;210:1117–24.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Zheng Y, Valdez PA, Danilenko DM, et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med. 2008;14:282–9.PubMedCrossRef Zheng Y, Valdez PA, Danilenko DM, et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med. 2008;14:282–9.PubMedCrossRef
32.
Zurück zum Zitat Tumanov AV, Koroleva EP, Guo X, et al. Lymphotoxin controls the IL-22 protection pathway in gut innate lymphoid cells during mucosal pathogen challenge. Cell Host Microbe. 2011;10:44–53.PubMedPubMedCentralCrossRef Tumanov AV, Koroleva EP, Guo X, et al. Lymphotoxin controls the IL-22 protection pathway in gut innate lymphoid cells during mucosal pathogen challenge. Cell Host Microbe. 2011;10:44–53.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Sonnenberg GF, Monticelli LA, Elloso MM, et al. CD4(+) lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity. 2011;34:122–34.PubMedCrossRef Sonnenberg GF, Monticelli LA, Elloso MM, et al. CD4(+) lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity. 2011;34:122–34.PubMedCrossRef
34.
Zurück zum Zitat Durant L, Watford WT, Ramos HL, et al. Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis. Immunity. 2010;32:605–15.PubMedPubMedCentralCrossRef Durant L, Watford WT, Ramos HL, et al. Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis. Immunity. 2010;32:605–15.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Takeda K, Clausen BE, Kaisho T, et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity. 1999;10:39–49.PubMedCrossRef Takeda K, Clausen BE, Kaisho T, et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity. 1999;10:39–49.PubMedCrossRef
36.
Zurück zum Zitat Pestka S, Krause CD, Sarkar D, et al. Interleukin-10 and related cytokines and receptors. Annu Rev Immunol. 2004;22:929–79.PubMedCrossRef Pestka S, Krause CD, Sarkar D, et al. Interleukin-10 and related cytokines and receptors. Annu Rev Immunol. 2004;22:929–79.PubMedCrossRef
37.
Zurück zum Zitat Nagalakshmi ML, Rascle A, Zurawski S, et al. Interleukin-22 activates STAT3 and induces IL-10 by colon epithelial cells. Int Immunopharmacol. 2004;4:679–91.PubMedCrossRef Nagalakshmi ML, Rascle A, Zurawski S, et al. Interleukin-22 activates STAT3 and induces IL-10 by colon epithelial cells. Int Immunopharmacol. 2004;4:679–91.PubMedCrossRef
38.
Zurück zum Zitat Hainzl E, Stockinger S, Rauch I, et al. Intestinal epithelial cell tyrosine kinase 2 transduces IL-22 signals to protect from acute colitis. J Immunol. 2015;195:5011–24.PubMedPubMedCentralCrossRef Hainzl E, Stockinger S, Rauch I, et al. Intestinal epithelial cell tyrosine kinase 2 transduces IL-22 signals to protect from acute colitis. J Immunol. 2015;195:5011–24.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Wolk K, Witte E, Hoffmann U, et al. IL-22 induces lipopolysaccharide-binding protein in hepatocytes: a potential systemic role of IL-22 in Crohn’s disease. J Immunol. 2007;178:5973–81.PubMedCrossRef Wolk K, Witte E, Hoffmann U, et al. IL-22 induces lipopolysaccharide-binding protein in hepatocytes: a potential systemic role of IL-22 in Crohn’s disease. J Immunol. 2007;178:5973–81.PubMedCrossRef
40.
Zurück zum Zitat Dumoutier L, de Meester C, Tavernier J, et al. New activation modus of STAT3: a tyrosine-less region of the interleukin-22 receptor recruits STAT3 by interacting with its coiled-coil domain. J Biol Chem. 2009;284:26377–84.PubMedPubMedCentralCrossRef Dumoutier L, de Meester C, Tavernier J, et al. New activation modus of STAT3: a tyrosine-less region of the interleukin-22 receptor recruits STAT3 by interacting with its coiled-coil domain. J Biol Chem. 2009;284:26377–84.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Wolk K, Witte E, Wallace E, et al. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur J Immunol. 2006;36:1309–23.PubMedCrossRef Wolk K, Witte E, Wallace E, et al. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur J Immunol. 2006;36:1309–23.PubMedCrossRef
42.
Zurück zum Zitat Liang SC, Tan XY, Luxenberg DP, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 2006;203:2271–9.PubMedPubMedCentralCrossRef Liang SC, Tan XY, Luxenberg DP, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 2006;203:2271–9.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Pham TA, Clare S, Goulding D, et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe. 2014;16:504–16.PubMedPubMedCentralCrossRef Pham TA, Clare S, Goulding D, et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe. 2014;16:504–16.PubMedPubMedCentralCrossRef
44.
45.
Zurück zum Zitat Mukherjee S, Zheng H, Derebe MG, et al. Antibacterial membrane attack by a pore-forming intestinal C-type lectin. Nature. 2014;505:103–7.PubMedCrossRef Mukherjee S, Zheng H, Derebe MG, et al. Antibacterial membrane attack by a pore-forming intestinal C-type lectin. Nature. 2014;505:103–7.PubMedCrossRef
46.
Zurück zum Zitat Brandl K, Plitas G, Schnabl B, et al. MyD88-mediated signals induce the bactericidal lectin RegIII gamma and protect mice against intestinal Listeria monocytogenes infection. J Exp Med. 2007;204:1891–900.PubMedPubMedCentralCrossRef Brandl K, Plitas G, Schnabl B, et al. MyD88-mediated signals induce the bactericidal lectin RegIII gamma and protect mice against intestinal Listeria monocytogenes infection. J Exp Med. 2007;204:1891–900.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Cox JH, Kljavin NM, Ota N, et al. Opposing consequences of IL-23 signaling mediated by innate and adaptive cells in chemically induced colitis in mice. Mucosal Immunol. 2012;5:99–109.PubMedCrossRef Cox JH, Kljavin NM, Ota N, et al. Opposing consequences of IL-23 signaling mediated by innate and adaptive cells in chemically induced colitis in mice. Mucosal Immunol. 2012;5:99–109.PubMedCrossRef
48.
Zurück zum Zitat Aden K, Rehman A, Falk-Paulsen M, et al. Epithelial IL-23R signaling licenses protective IL-22 responses in intestinal inflammation. Cell Rep. 2016;16:2208–18.PubMedPubMedCentralCrossRef Aden K, Rehman A, Falk-Paulsen M, et al. Epithelial IL-23R signaling licenses protective IL-22 responses in intestinal inflammation. Cell Rep. 2016;16:2208–18.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Ikhtaire S, Shajib MS, Reinisch W, et al. Fecal calprotectin: its scope and utility in the management of inflammatory bowel disease. J Gastroenterol. 2016;51:434–46.PubMedCrossRef Ikhtaire S, Shajib MS, Reinisch W, et al. Fecal calprotectin: its scope and utility in the management of inflammatory bowel disease. J Gastroenterol. 2016;51:434–46.PubMedCrossRef
50.
Zurück zum Zitat Lapeire L, Hendrix A, Lambein K, et al. Cancer-associated adipose tissue promotes breast cancer progression by paracrine oncostatin M and Jak/STAT3 signaling. Cancer Res. 2014;74:6806–19.PubMedCrossRef Lapeire L, Hendrix A, Lambein K, et al. Cancer-associated adipose tissue promotes breast cancer progression by paracrine oncostatin M and Jak/STAT3 signaling. Cancer Res. 2014;74:6806–19.PubMedCrossRef
51.
Zurück zum Zitat Hattrup CL, Gendler SJ. Structure and function of the cell surface (tethered) mucins. Annu Rev Physiol. 2008;70:431–57.PubMedCrossRef Hattrup CL, Gendler SJ. Structure and function of the cell surface (tethered) mucins. Annu Rev Physiol. 2008;70:431–57.PubMedCrossRef
52.
53.
Zurück zum Zitat Nishida A, Lau CW, Zhang M, et al. The membrane-bound mucin muc1 regulates Th17-cell responses and colitis in mice. Gastroenterology. 2012;142:865–74.PubMedCrossRef Nishida A, Lau CW, Zhang M, et al. The membrane-bound mucin muc1 regulates Th17-cell responses and colitis in mice. Gastroenterology. 2012;142:865–74.PubMedCrossRef
54.
Zurück zum Zitat Andoh A, Shioya M, Nishida A, et al. Expression of IL-24, an activator of the JAK1/STAT3/SOCS3 cascade, is enhanced in inflammatory bowel disease. J Immunol. 2009;183:687–95.PubMedCrossRef Andoh A, Shioya M, Nishida A, et al. Expression of IL-24, an activator of the JAK1/STAT3/SOCS3 cascade, is enhanced in inflammatory bowel disease. J Immunol. 2009;183:687–95.PubMedCrossRef
55.
Zurück zum Zitat Yuan ZL, Guan YJ, Wang L, et al. Central role of the threonine residue within the p + 1 loop of receptor tyrosine kinase in STAT3 constitutive phosphorylation in metastatic cancer cells. Mol Cell Biol. 2004;24:9390–400.PubMedPubMedCentralCrossRef Yuan ZL, Guan YJ, Wang L, et al. Central role of the threonine residue within the p + 1 loop of receptor tyrosine kinase in STAT3 constitutive phosphorylation in metastatic cancer cells. Mol Cell Biol. 2004;24:9390–400.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Begue B, Verdier J, Rieux-Laucat F, et al. Defective IL10 signaling defining a subgroup of patients with inflammatory bowel disease. Am J Gastroenterol. 2011;106:1544–55.PubMedCrossRef Begue B, Verdier J, Rieux-Laucat F, et al. Defective IL10 signaling defining a subgroup of patients with inflammatory bowel disease. Am J Gastroenterol. 2011;106:1544–55.PubMedCrossRef
57.
Zurück zum Zitat Kadayakkara DK, Beatty PL, Turner MS, et al. Inflammation driven by overexpression of the hypoglycosylated abnormal mucin 1 (MUC1) links inflammatory bowel disease and pancreatitis. Pancreas. 2010;39:510–5.PubMedPubMedCentralCrossRef Kadayakkara DK, Beatty PL, Turner MS, et al. Inflammation driven by overexpression of the hypoglycosylated abnormal mucin 1 (MUC1) links inflammatory bowel disease and pancreatitis. Pancreas. 2010;39:510–5.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Beatty PL, Plevy SE, Sepulveda AR, et al. Transgenic expression of human MUC1 in IL-10-/- mice accelerates inflammatory bowel disease and progression to colon cancer. J Immunol. 2007;179:735–9.PubMedCrossRef Beatty PL, Plevy SE, Sepulveda AR, et al. Transgenic expression of human MUC1 in IL-10-/- mice accelerates inflammatory bowel disease and progression to colon cancer. J Immunol. 2007;179:735–9.PubMedCrossRef
59.
60.
Zurück zum Zitat Neufert C, Pickert G, Zheng Y, et al. Activation of epithelial STAT3 regulates intestinal homeostasis. Cell Cycle. 2010;9:652–5.PubMedCrossRef Neufert C, Pickert G, Zheng Y, et al. Activation of epithelial STAT3 regulates intestinal homeostasis. Cell Cycle. 2010;9:652–5.PubMedCrossRef
61.
Zurück zum Zitat Fukui H, Sekikawa A, Tanaka H, et al. DMBT1 is a novel gene induced by IL-22 in ulcerative colitis. Inflamm Bowel Dis. 2011;17:1177–88.PubMedCrossRef Fukui H, Sekikawa A, Tanaka H, et al. DMBT1 is a novel gene induced by IL-22 in ulcerative colitis. Inflamm Bowel Dis. 2011;17:1177–88.PubMedCrossRef
62.
Zurück zum Zitat Lindemans CA, Calafiore M, Mertelsmann AM, et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528:560–4.PubMedPubMedCentralCrossRef Lindemans CA, Calafiore M, Mertelsmann AM, et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528:560–4.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Aparicio-Domingo P, Romera-Hernandez M, Karrich JJ, et al. Type 3 innate lymphoid cells maintain intestinal epithelial stem cells after tissue damage. J Exp Med. 2015;212:1783–91.PubMedPubMedCentralCrossRef Aparicio-Domingo P, Romera-Hernandez M, Karrich JJ, et al. Type 3 innate lymphoid cells maintain intestinal epithelial stem cells after tissue damage. J Exp Med. 2015;212:1783–91.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Bootz F, Ziffels B, Neri D. Antibody-based targeted delivery of interleukin-22 promotes rapid clinical recovery in mice with DSS-induced colitis. Inflamm Bowel Dis. 2016;22:2098–105.PubMedCrossRef Bootz F, Ziffels B, Neri D. Antibody-based targeted delivery of interleukin-22 promotes rapid clinical recovery in mice with DSS-induced colitis. Inflamm Bowel Dis. 2016;22:2098–105.PubMedCrossRef
65.
Zurück zum Zitat Monteleone I, Rizzo A, Sarra M, et al. Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract. Gastroenterology. 2011;141:237–48.PubMedCrossRef Monteleone I, Rizzo A, Sarra M, et al. Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract. Gastroenterology. 2011;141:237–48.PubMedCrossRef
67.
Zurück zum Zitat Tsai PY, Zhang B, He WQ, et al. IL-22 upregulates epithelial claudin-2 to drive diarrhea and enteric pathogen clearance. Cell Host Microbe. 2017;21:671–81.PubMedCrossRef Tsai PY, Zhang B, He WQ, et al. IL-22 upregulates epithelial claudin-2 to drive diarrhea and enteric pathogen clearance. Cell Host Microbe. 2017;21:671–81.PubMedCrossRef
68.
Zurück zum Zitat Jinnohara T, Kanaya T, Hase K, et al. IL-22BP dictates characteristics of Peyer’s patch follicle-associated epithelium for antigen uptake. J Exp Med. 2017;214:1607–18.PubMedPubMedCentralCrossRef Jinnohara T, Kanaya T, Hase K, et al. IL-22BP dictates characteristics of Peyer’s patch follicle-associated epithelium for antigen uptake. J Exp Med. 2017;214:1607–18.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Gulhane M, Murray L, Lourie R, et al. High fat diets induce colonic epithelial cell stress and inflammation that is reversed by IL-22. Sci Rep. 2016;6:28990.PubMedPubMedCentralCrossRef Gulhane M, Murray L, Lourie R, et al. High fat diets induce colonic epithelial cell stress and inflammation that is reversed by IL-22. Sci Rep. 2016;6:28990.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Qiu J, Heller JJ, Guo X, et al. The Aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity. 2012;36:92–104.PubMedCrossRef Qiu J, Heller JJ, Guo X, et al. The Aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity. 2012;36:92–104.PubMedCrossRef
71.
Zurück zum Zitat Schulz SM, Köhler G, Schütze N, et al. Protective immunity to systemic infection with attenuated Salmonella enterica serovar enteritidis in the absence of IL-12 is associated with IL-23-dependent IL-22, but not IL-17. J Immunol. 2008;181:7891–901.PubMedCrossRef Schulz SM, Köhler G, Schütze N, et al. Protective immunity to systemic infection with attenuated Salmonella enterica serovar enteritidis in the absence of IL-12 is associated with IL-23-dependent IL-22, but not IL-17. J Immunol. 2008;181:7891–901.PubMedCrossRef
72.
Zurück zum Zitat De Luca A, Zelante T, D’Angelo C, et al. IL-22 defines a novel immune pathway of antifungal resistance. Mucosal Immunol. 2010;3:361–73.PubMedCrossRef De Luca A, Zelante T, D’Angelo C, et al. IL-22 defines a novel immune pathway of antifungal resistance. Mucosal Immunol. 2010;3:361–73.PubMedCrossRef
73.
Zurück zum Zitat Kamanaka M, Huber S, Zenewicz LA, et al. Memory/effector (CD45RB(lo)) CD4 T cells are controlled directly by IL-10 and cause IL-22-dependent intestinal pathology. J Exp Med. 2011;208:1027–40.PubMedPubMedCentralCrossRef Kamanaka M, Huber S, Zenewicz LA, et al. Memory/effector (CD45RB(lo)) CD4 T cells are controlled directly by IL-10 and cause IL-22-dependent intestinal pathology. J Exp Med. 2011;208:1027–40.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Eken A, Singh AK, Treuting PM, et al. IL-23R+ innate lymphoid cells induce colitis via interleukin-22-dependent mechanism. Mucosal Immunol. 2014;7:143–54.PubMedCrossRef Eken A, Singh AK, Treuting PM, et al. IL-23R+ innate lymphoid cells induce colitis via interleukin-22-dependent mechanism. Mucosal Immunol. 2014;7:143–54.PubMedCrossRef
75.
Zurück zum Zitat Wolk K, Haugen HS, Xu W, et al. IL-22 and IL-20 are key mediators of the epidermal alterations in psoriasis while IL-17 and IFN-gamma are not. J Mol Med. 2009;87:523–36.PubMedCrossRef Wolk K, Haugen HS, Xu W, et al. IL-22 and IL-20 are key mediators of the epidermal alterations in psoriasis while IL-17 and IFN-gamma are not. J Mol Med. 2009;87:523–36.PubMedCrossRef
76.
Zurück zum Zitat Sonnenberg GF, Nair MG, Kirn TJ, et al. Pathological versus protective functions of IL-22 in airway inflammation are regulated by IL-17A. J Exp Med. 2010;207:1293–305.PubMedPubMedCentralCrossRef Sonnenberg GF, Nair MG, Kirn TJ, et al. Pathological versus protective functions of IL-22 in airway inflammation are regulated by IL-17A. J Exp Med. 2010;207:1293–305.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Krausgruber T, Schiering C, Adelmann K, et al. T-bet is a key modulator of IL-23-driven pathogenic CD4(+) T cell responses in the intestine. Nat Commun. 2016;7:11627.PubMedPubMedCentralCrossRef Krausgruber T, Schiering C, Adelmann K, et al. T-bet is a key modulator of IL-23-driven pathogenic CD4(+) T cell responses in the intestine. Nat Commun. 2016;7:11627.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Moriwaki K, Balaji S, McQuade T, et al. The necroptosis adaptor RIPK3 promotes injury-induced cytokine expression and tissue repair. Immunity. 2014;41:567–78.PubMedPubMedCentralCrossRef Moriwaki K, Balaji S, McQuade T, et al. The necroptosis adaptor RIPK3 promotes injury-induced cytokine expression and tissue repair. Immunity. 2014;41:567–78.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Liu X, Wei W, Li X, et al. BMI1 and MEL18 Promote Colitis-Associated Cancer in Mice via REG3B and STAT3. Gastroenterology. 2017 Aug 2. pii: S0016-5085(17)35976-0 (In Press). Liu X, Wei W, Li X, et al. BMI1 and MEL18 Promote Colitis-Associated Cancer in Mice via REG3B and STAT3. Gastroenterology. 2017 Aug 2. pii: S0016-5085(17)35976-0 (In Press).
80.
Zurück zum Zitat Xu W, Presnell SR, Parrish-Novak J, et al. A soluble class II cytokine receptor, IL-22RA2, is a naturally occurring IL-22 antagonist. Proc Natl Acad Sci USA. 2001;98:9511–6.PubMedPubMedCentralCrossRef Xu W, Presnell SR, Parrish-Novak J, et al. A soluble class II cytokine receptor, IL-22RA2, is a naturally occurring IL-22 antagonist. Proc Natl Acad Sci USA. 2001;98:9511–6.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Dumoutier L, Lejeune D, Colau D, et al. Cloning and characterization of IL-22 binding protein, a natural antagonist of IL-10-related T cell-derived inducible factor/IL-22. J Immunol. 2001;166:7090–5.PubMedCrossRef Dumoutier L, Lejeune D, Colau D, et al. Cloning and characterization of IL-22 binding protein, a natural antagonist of IL-10-related T cell-derived inducible factor/IL-22. J Immunol. 2001;166:7090–5.PubMedCrossRef
82.
Zurück zum Zitat Kotenko SV, Izotova LS, Mirochnitchenko OV, et al. Identification, cloning, and characterization of a novel soluble receptor that binds IL-22 and neutralizes its activity. J Immunol. 2001;166:7096–103.PubMedCrossRef Kotenko SV, Izotova LS, Mirochnitchenko OV, et al. Identification, cloning, and characterization of a novel soluble receptor that binds IL-22 and neutralizes its activity. J Immunol. 2001;166:7096–103.PubMedCrossRef
83.
Zurück zum Zitat Huber S, Gagliani N, Zenewicz LA, et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. 2012;491:259–63.PubMedPubMedCentralCrossRef Huber S, Gagliani N, Zenewicz LA, et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. 2012;491:259–63.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Pelczar P, Witkowski M, Perez LG, et al. A pathogenic role for T cell-derived IL-22BP in inflammatory bowel disease. Science. 2016;354:358–62.PubMedCrossRef Pelczar P, Witkowski M, Perez LG, et al. A pathogenic role for T cell-derived IL-22BP in inflammatory bowel disease. Science. 2016;354:358–62.PubMedCrossRef
85.
Zurück zum Zitat Ratsimandresy RA, Indramohan M, Dorfleutner A, et al. The AIM2 inflammasome is a central regulator of intestinal homeostasis through the IL-18/IL-22/STAT3 pathway. Cell Mol Immunol. 2017;14:127–42.PubMedCrossRef Ratsimandresy RA, Indramohan M, Dorfleutner A, et al. The AIM2 inflammasome is a central regulator of intestinal homeostasis through the IL-18/IL-22/STAT3 pathway. Cell Mol Immunol. 2017;14:127–42.PubMedCrossRef
86.
Zurück zum Zitat Martin JC, Bériou G, Heslan H, et al. Interleukin-22 binding protein (IL-22BP) is constitutively expressed by a subset of conventional dendritic cells and is strongly induced by retinoic acid. Mucosal Immunol. 2014;7:101–13.PubMedCrossRef Martin JC, Bériou G, Heslan H, et al. Interleukin-22 binding protein (IL-22BP) is constitutively expressed by a subset of conventional dendritic cells and is strongly induced by retinoic acid. Mucosal Immunol. 2014;7:101–13.PubMedCrossRef
87.
Zurück zum Zitat Martin JC, Bériou G, Heslan M, et al. IL-22BP is produced by eosinophils in human gut and blocks IL-22 protective actions during colitis. Mucosal Immunol. 2016;9:539–49.PubMedCrossRef Martin JC, Bériou G, Heslan M, et al. IL-22BP is produced by eosinophils in human gut and blocks IL-22 protective actions during colitis. Mucosal Immunol. 2016;9:539–49.PubMedCrossRef
88.
Zurück zum Zitat Lim C, Hong M, Savan R. Human IL-22 binding protein isoforms act as a rheostat for IL-22 signaling. Sci Signal. 2016;9:ra95.PubMedCrossRef Lim C, Hong M, Savan R. Human IL-22 binding protein isoforms act as a rheostat for IL-22 signaling. Sci Signal. 2016;9:ra95.PubMedCrossRef
91.
Zurück zum Zitat Kakuta Y, Kimura T, Negoro K, et al. Increased expression of IL12B mRNA transcribed from the risk haplotype for Crohn’s disease is a risk factor for disease relapse in Japanese patients. J Gastroenterol. 2017;. doi:10.1007/s00535-017-1322-5.PubMedCentral Kakuta Y, Kimura T, Negoro K, et al. Increased expression of IL12B mRNA transcribed from the risk haplotype for Crohn’s disease is a risk factor for disease relapse in Japanese patients. J Gastroenterol. 2017;. doi:10.​1007/​s00535-017-1322-5.PubMedCentral
92.
Zurück zum Zitat Glocker EO, Kotlarz D, Boztug K, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361:2033–45.PubMedPubMedCentralCrossRef Glocker EO, Kotlarz D, Boztug K, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361:2033–45.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Silverberg MS, Cho JH, Rioux JD, et al. Ulcerative colitis-risk loci on chromosomes 1p36 and 12q15 found by genome-wide association study. Nat Genet. 2009;41:216–20.PubMedPubMedCentralCrossRef Silverberg MS, Cho JH, Rioux JD, et al. Ulcerative colitis-risk loci on chromosomes 1p36 and 12q15 found by genome-wide association study. Nat Genet. 2009;41:216–20.PubMedPubMedCentralCrossRef
94.
95.
Zurück zum Zitat Yamamoto-Furusho JK, Sánchez-Morales GE, García-Rangel D, et al. Genetic polymorphisms of interleukin-22 in patients with ulcerative colitis. Rev Gastroenterol Mex. 2016;81:86–90.PubMed Yamamoto-Furusho JK, Sánchez-Morales GE, García-Rangel D, et al. Genetic polymorphisms of interleukin-22 in patients with ulcerative colitis. Rev Gastroenterol Mex. 2016;81:86–90.PubMed
96.
Zurück zum Zitat Franke A, McGovern DP, Barrett JC, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet. 2010;42:1118–25.PubMedPubMedCentralCrossRef Franke A, McGovern DP, Barrett JC, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet. 2010;42:1118–25.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Kaplan GG, Ng SC. Understanding and preventing the global increase of inflammatory bowel disease. Gastroenterology. 2017;152:313–21.PubMedCrossRef Kaplan GG, Ng SC. Understanding and preventing the global increase of inflammatory bowel disease. Gastroenterology. 2017;152:313–21.PubMedCrossRef
98.
Zurück zum Zitat Colombel JF, Watson AJ, Neurath MF. The 10 remaining mysteries of inflammatory bowel disease. Gut. 2008;57:429–33.PubMedCrossRef Colombel JF, Watson AJ, Neurath MF. The 10 remaining mysteries of inflammatory bowel disease. Gut. 2008;57:429–33.PubMedCrossRef
99.
Zurück zum Zitat Monteleone I, Pallone F, Monteleone G. Aryl hydrocarbon receptor and colitis. Semin Immunopathol. 2013;35:671–5.PubMedCrossRef Monteleone I, Pallone F, Monteleone G. Aryl hydrocarbon receptor and colitis. Semin Immunopathol. 2013;35:671–5.PubMedCrossRef
100.
Zurück zum Zitat Iqbal J, Sun L, Cao J, et al. Smoke carcinogens cause bone loss through the aryl hydrocarbon receptor and induction of Cyp1 enzymes. Proc Natl Acad Sci USA. 2013;110:11115–20.PubMedPubMedCentralCrossRef Iqbal J, Sun L, Cao J, et al. Smoke carcinogens cause bone loss through the aryl hydrocarbon receptor and induction of Cyp1 enzymes. Proc Natl Acad Sci USA. 2013;110:11115–20.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Xue J, Zhao Q, Sharma V, et al. Aryl hydrocarbon receptor ligands in cigarette smoke induce production of interleukin-22 to promote pancreatic fibrosis in models of chronic pancreatitis. Gastroenterology. 2016;151:1206–17.PubMedPubMedCentralCrossRef Xue J, Zhao Q, Sharma V, et al. Aryl hydrocarbon receptor ligands in cigarette smoke induce production of interleukin-22 to promote pancreatic fibrosis in models of chronic pancreatitis. Gastroenterology. 2016;151:1206–17.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Zelante T, Iannitti RG, Cunha C, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39:372–85.PubMedCrossRef Zelante T, Iannitti RG, Cunha C, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39:372–85.PubMedCrossRef
103.
Zurück zum Zitat Lamas B, Richard ML, Leducq V, et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med. 2016;22:598–605.PubMedPubMedCentralCrossRef Lamas B, Richard ML, Leducq V, et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med. 2016;22:598–605.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Sugimoto S, Naganuma M, Kanai T. Indole compounds may be promising medicines for ulcerative colitis. J Gastroenterol. 2016;51:853–61.PubMedCrossRef Sugimoto S, Naganuma M, Kanai T. Indole compounds may be promising medicines for ulcerative colitis. J Gastroenterol. 2016;51:853–61.PubMedCrossRef
105.
Zurück zum Zitat Sugimoto S, Naganuma M, Kiyohara H, et al. Clinical efficacy and safety of oral qing-dai in patients with ulcerative colitis: a single-center open-label prospective study. Digestion. 2016;93:193–201.PubMedCrossRef Sugimoto S, Naganuma M, Kiyohara H, et al. Clinical efficacy and safety of oral qing-dai in patients with ulcerative colitis: a single-center open-label prospective study. Digestion. 2016;93:193–201.PubMedCrossRef
106.
Zurück zum Zitat Kawai S, Iijima H, Shinzaki S, et al. Indigo naturalis ameliorates murine dextran sodium sulfate-induced colitis via aryl hydrocarbon receptor activation. J Gastroenterol. 2017;52:904–19.PubMedCrossRef Kawai S, Iijima H, Shinzaki S, et al. Indigo naturalis ameliorates murine dextran sodium sulfate-induced colitis via aryl hydrocarbon receptor activation. J Gastroenterol. 2017;52:904–19.PubMedCrossRef
107.
Zurück zum Zitat Chebli K, Papon L, Paul C, et al. The anti-HIV candidate Abx464 dampens intestinal inflammation by triggering Il-22 production in activated macrophages. Sci Rep. 2017;7:4860.PubMedPubMedCentralCrossRef Chebli K, Papon L, Paul C, et al. The anti-HIV candidate Abx464 dampens intestinal inflammation by triggering Il-22 production in activated macrophages. Sci Rep. 2017;7:4860.PubMedPubMedCentralCrossRef
108.
109.
Zurück zum Zitat Bollrath J, Phesse TJ, von Burstin VA, et al. gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis. Cancer Cell. 2009;15:91–102.PubMedCrossRef Bollrath J, Phesse TJ, von Burstin VA, et al. gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis. Cancer Cell. 2009;15:91–102.PubMedCrossRef
Metadaten
Titel
Clinical importance of IL-22 cascade in IBD
verfasst von
Atsushi Mizoguchi
Arisa Yano
Hidetomo Himuro
Yui Ezaki
Takayuki Sadanaga
Emiko Mizoguchi
Publikationsdatum
26.10.2017
Verlag
Springer Japan
Erschienen in
Journal of Gastroenterology / Ausgabe 4/2018
Print ISSN: 0944-1174
Elektronische ISSN: 1435-5922
DOI
https://doi.org/10.1007/s00535-017-1401-7

Weitere Artikel der Ausgabe 4/2018

Journal of Gastroenterology 4/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.