Abstract

Interaction between intestinal epithelial cells (IECs) and the underlying immune systems is critical for maintaining intestinal immune homeostasis and mounting appropriate immune responses. We have previously showed that the T helper type 1 (TH1) cytokine IL-12 plays a key role in the delicate immunological balance in the gut and the lack of appropriate levels of IL-12 had important consequences for health and disease, particularly with regard to food allergy. Here, we sought to understand the role of IL-12 in the regulation of lymphoepithelial cross talk and how this interaction affects immune responses locally and systemically. Using a combination of microscopy and flow cytometry techniques we observed that freshly isolated IECs expressed an incomplete, yet functional IL-12 receptor (IL-12R) formed solely by the IL-12Rβ2 chain that albeit the lack of the complementary IL-12β1 chain responded to ex vivo challenge with IL-12. Furthermore, the expression of IL-12Rβ2 on IECs is strategically located at the interface between epithelial and immune cells of the lamina propria and using in vitro coculture models and primary intestinal organoids we showed that immune-derived signals were required for the expression of IL-12Rβ2 on IECs. The biological relevance of the IEC-associated IL-12Rβ2 was assessed in vivo in a mouse model of food allergy characterized by allergy-associated diminished intestinal levels of IL-12 and in chimeric mice that lack the IL-12Rβ2 chain on IECs. These experimental models enabled us to show that the antiallergic properties of orally delivered recombinant Lactococcus lactis secreting bioactive IL-12 (rLc-IL12) were reduced in mice lacking the IL-12β2 chain on IECs. Finally, we observed that the oral delivery of IL-12 was accompanied by the downregulation of the production of the IEC-derived proallergic cytokine thymic stromal lymphopoietin (TSLP). However, further analysis of intestinal levels of TSLP in IL-12Rβ2−/− mice suggested that this event was not directly linked to the IEC-associated IL-12Rβ2 chain. We interpreted these data as showing that IEC-associated IL12Rβ2 is a component of the cytokine network operating at the interface between the intestinal epithelium and immune system that plays a role in immune regulation.

Highlights

  • The constant dialog between the intestinal epithelium and under­ lying immune system plays a key role in intestinal immune homeostasis by shaping the appropriate immunological microenvi­ ronment [1]

  • The TH1 cytokine IL-12 binds to a heterodimeric IL-12Rβ1/β2 complexes on a variety of immune cells [21]

  • We report that mouse intestinal epithelial cells (IECs) expressed the IL-12Rβ2 but not the complementary IL-12Rβ1 chain that functions as an incom­ plete, yet functional IL-12 receptor (IL-12R) that plays a role in lympho-epithelial cross talk

Read more

Summary

Introduction

The constant dialog between the intestinal epithelium and under­ lying immune system plays a key role in intestinal immune homeostasis by shaping the appropriate immunological microenvi­ ronment [1]. The T helper type 1 (TH1) cytokine IL-12 plays a variety of roles in intestinal immunity ranging from response to pathogens [8], interaction with group 1 innate lymphoid cells in maintaining barrier integrity [9] to protecting against food allergy [10]. IEC-associated IL-12Rβ2 operates as an incomplete, yet still operational IL12R the expression of which is under the control of immune cell-derived signals This observation prompted us to test the hypothesis that the presence of IEC-associated IL-12Rβ2 could directly affect the regulatory properties of IECs. This observation prompted us to test the hypothesis that the presence of IEC-associated IL-12Rβ2 could directly affect the regulatory properties of IECs This was done by adopting a strategy based on oral administration of a Lactococcus lactis strain genetically engineered to produce biologically active IL-12 (rLc-IL12) [12] in allergic wild-type (wt) and chimeric mice that lacked IL-12Rβ2 on the intestinal epithelium. IL-12 mediated suppression of food allergy was accompanied by the downregulation of the IEC-derived cytokine TSLP

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call