Abstract

Dendritic cells (DCs) are key mediators of transplant rejection. Numerous factors have been identified that regulate transplant immunopathology by modulating the function of DCs. Among these, microRNAs (miRNAs), small non-coding RNA molecules, have received much attention. The miRNA miR-223 is very highly expressed and tightly regulated in hematopoietic cells. It plays an important role in modulating the immune response by regulating neutrophils and macrophages, and its dysregulation contributes to multiple types of immune diseases. However, the role of miR-223 in immune rejection is unclear. Here, we observed expression of miR-223 in patients and mice who had undergone heart transplantation and found that it increased in the serum of both, and also in DCs from the spleens of recipient mice, although it was unchanged in splenic T cells. We also found that miR-223 expression decreased in lipopolysaccharide-stimulated DCs. Increasing the level of miR-223 in DCs promoted polarization of DCs toward a tolerogenic phenotype, which indicates that miR-223 can attenuate activation and maturation of DCs. MiR-223 effectively induced regulatory T cells (Tregs) by inhibiting the function of antigen-presenting DCs. In addition, we identified Irak1 as a miR-223 target gene and an essential regulator of DC maturation. In mouse allogeneic heterotopic heart transplantation models, grafts survived longer and suffered less immune cell infiltration in mice with miR-223-overexpressing immature (im)DCs. In the miR-223-overexpressing imDC recipients, T cells from spleen differentiated into Tregs, and the level of IL-10 in heart grafts was markedly higher than that in the control group. In conclusion, miR-223 regulates the function of DCs via Irak1, differentiation of T cells into Tregs, and secretion of IL-10, thereby suppressing allogeneic heart graft rejection.

Highlights

  • Heart transplantation is currently the most effective treatment for end-stage heart failure

  • The results showed that expression of CD80, CD86, and major histocompatibility complex (MHC)-II was unchanged in the three groups (Figures 3B, C), which indicated that miR-NC had no impact on the bone marrow–derived DC (BMDC) phenotype

  • We found that imBMDCs and miR-NC imBMDCs expressed high levels of MHC-II, CD86, and CD80 upon LPS stimulation, whereas the miR-223 mimic-imBMDCs retained low levels of these maturation markers (Figures 3B, C)

Read more

Summary

Introduction

Heart transplantation is currently the most effective treatment for end-stage heart failure. There is an urgent need for an improved approach to treatment that can induce allograftspecific tolerance in the recipient and enable long-term graft survival while avoiding the use of immunosuppression that causes toxic side effects. DCs are essential for the induction of an immune response to the graft or in maintaining immunological tolerance, depending on their activation state and maturation status [2], for which they are classified as either mature (mDCs) or immature (imDCs). Immature DCs (imDCs) are induced by numerous immunosuppressive agents, including cytokines (IL-10, TGF-b) and endogenous immunosuppressants (glucocorticoids) [8] They play important roles in inducing peripheral tolerance via specific mechanisms, including induction of Tregs, promotion of anti-inflammatory cytokine secretion [9], suppression of effector T cells, and negative modulation of Th1/Th2 immune responses [10]. Dynamic regulation of DC maturation is crucial for influencing the balance between tolerance and immunity in T-cells

Objectives
Methods
Results
Discussion
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