Abstract

BackgroundIn a previous study, activation of the peroxisome proliferator–activated receptor γ (PPARγ) inhibited chronic cardiac rejection. However, because of the complexity of chronic rejection and the fact that PPARγ is widely expressed in immune cells, the mechanism of the PPARγ - induced protective effect was unclear.Materials and MethodsA chronic rejection model was established using B6.C-H-2bm12KhEg (H-2bm12) mice as donors, and MHC II-mismatched T-cell-specific PPARγ knockout mice or wild type (WT) littermates as recipients. The allograft lesion was assessed by histology and immunohistochemistry. T cells infiltrates in the allograft were isolated, and cytokines and subpopulations were detected using cytokine arrays and flow cytometry. Transcription levels in the allograft were measured by RT-PCR. In vitro, the T cell subset differentiation was investigated after culture in various polarizing conditions. PPARγ-deficient regularory T cells (Treg) were cocultured with monocytes to test their ability to induce alternatively activated macrophages (AAM).ResultsT cell-specific PPARγ knockout recipients displayed reduced cardiac allograft survival and an increased degree of pathology compared with WT littermates. T cell-specific PPARγ knockout resulted in more CD4+ T cells infiltrating into the allograft and altered the Th1/Th2 and Th17/Treg ratios. The polarization of AAM was also reduced by PPARγ deficiency in T cells through the action of Th2 and Treg. PPARγ-deficient T cells eliminated the pioglitazone-induced polarization of AAM and reduced allograft survival.ConclusionsPPARγ-deficient T cells influenced the T cell subset and AAM polarization in chronic allograft rejection. The mechanism of PPARγ activation in transplantation tolerance could yield a novel treatment without side effects.

Highlights

  • In end-stage heart disease, heart transplantation is becoming the most important clinical treatment [1]

  • T cell-specific peroxisome proliferator–activated receptor c (PPARc) knockout recipients displayed reduced cardiac allograft survival and an increased degree of pathology compared with wild type (WT) littermates

  • PPARc-deficient T cells eliminated the pioglitazone-induced polarization of activated macrophage (AAM) and reduced allograft survival

Read more

Summary

Introduction

In end-stage heart disease, heart transplantation is becoming the most important clinical treatment [1]. Previous studies have demonstrated that an immune mechanism participates in chronic allograft rejection. Many studies have focused on CD4+ T helper cells and their subsets, such as Th1, Th2, Th17 and regulatory T cells (Treg), in the process of chronic allograft rejection. Within the complex mechanism of chronic allograft rejection, both T cells and macrophages participate in the lesion of allografts [3]. These cells are immunotherapy targets in chronic allograft rejection. Because of the complexity of chronic rejection and the fact that PPARc is widely expressed in immune cells, the mechanism of the PPARc - induced protective effect was unclear

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.