Abstract

Experimental autoimmune uveoretinitis (EAU) is an organ-specific T cell-mediated disease induced by immunizing mice with interphotoreceptor retinoid binding protein (IRBP). Autoaggressive CD4+ T cells are the major pathogenic population for EAU. We investigated the contribution of Notch signaling in T cells to EAU pathogenesis because Notch signaling regulates various aspects of CD4+ T cell functions. Rbpj is required for Notch signaling, and Rbpj deficiency in T cells inhibited EAU disease severity. The amelioration of EAU in T cell-specific Rbpj-deficient mice correlated with low levels of IL-22 production from CD4+ T cells, although IRBP-specific CD4+ T cell proliferation and Th17 differentiation were unaffected. Administration of recombinant IL-22 during the late phase, but not the early phase, of EAU increased EAU clinical scores in T cell-specific Rbpj-deficient mice. Notch inhibition in mice immunized with IRBP with a γ-secretase inhibitor (GSI) suppressed EAU progression, even when GSI was administered as late as 13 days after IRBP immunization. Our data demonstrate that Rbpj/Notch-mediated IL-22 production in T cells has a key pathological role in the late phase of EAU, and suggest that Notch blockade might be a useful therapeutic approach for treating EAU.

Highlights

  • Experimental autoimmune uveoretinitis (EAU) is an organspecific, T cell-mediated disease initiated by immunizing mice with retinal antigens or their fragments [1] [2] [3] [4]

  • IL-22 acted as an inflammatory cytokine during the late phase but not the early phase of EAU progression

  • The present study showed that administration of Recombinant IL-22 (rIL-22) during the late phase aggravated EAU in Rbpjf/f-CD4 mice, while clinical scores were unaffected when rIL-22 was administered only during the early phase

Read more

Summary

Introduction

Experimental autoimmune uveoretinitis (EAU) is an organspecific, T cell-mediated disease initiated by immunizing mice with retinal antigens or their fragments [1] [2] [3] [4]. EAU represents a breakdown in tolerance to immunologically privileged retinal antigens, such as interphotoreceptor retinoid binding protein (IRBP) and arrestin, which function in the visual cycle. Th1 cells are activated in EAU, it has been reported that IL-12 down regulates EAU, and treatment of mice with EAU with anti-IFN-c antibodies aggravated the disease [6] [7]. It is essential to identify which T cells or which cytokines are crucial for each phase during EAU progression or human autoimmune uveoretinitis

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