4.8 Article

Mechanistic contributions of Kupffer cells and liver sinusoidal endothelial cells in nanoparticle-induced antigen-specific immune tolerance

期刊

BIOMATERIALS
卷 283, 期 -, 页码 -

出版社

ELSEVIER SCI LTD
DOI: 10.1016/j.biomaterials.2022.121457

关键词

PLG; Nanoparticles; Immune tolerance; Immunomodulation; Kupffer cells; Liver sinusoidal endothelial cells

资金

  1. NIH [EB-013198, T32GM007863]
  2. University of Michigan Rackham Predoctoral Fellowship
  3. National Institute of Allergy and Infectious Diseases of the National Institutes of Health [R01AI155678, R01AI148076]

向作者/读者索取更多资源

The delivery of disease-relevant antigens by polymeric nanoparticles has shown to induce immune tolerance in various disorders. This study investigated the contribution of liver sinusoidal endothelial cells and Kupffer cells in nanoparticle-induced tolerance in a mouse model of multiple sclerosis. The results suggest that both cell types play important roles in the induction of tolerance through the secretion of specific factors and interaction with CD4 T cells. This mechanistic support provides insights for the development of biomaterial platforms in clinical trials.
The intravenous delivery of disease-relevant antigens (Ag) by polymeric nanoparticles (NP-Ags) has demonstrated Ag-specific immune tolerance in autoimmune and allergic disorders as well as allogeneic transplant rejection. NP-Ags are observed to distribute to the spleen, which has an established role in the induction of immune tolerance. However, studies have shown that the spleen is dispensable for NP-Ag-induced tolerance, suggesting significant contributions from other immunological sites. Here, we investigated the tolerogenic contributions of Kupffer cells (KCs) and liver sinusoidal endothelial cells (LSECs) to NP-Ag-induced tolerance in a mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Intravenously delivered Ag-conjugated poly(lactide-co-glycolide) NPs (PLG-Ag) distributed largely to the liver, where they associated with both KCs and LSECs. This distribution was accompanied by CD4 T cell accumulation, clonal deletion, and PD-L1 expression by KCs and LSECs. Ex vivo co-cultures of PLG-Ag-treated KCs or LSECs with Ag-specific CD4 T cells resulted in PGE(2) and IL-10 or PGE(2) secretion, respectively. KC depletion and adoptive transfer experiments demonstrated that KCs were sufficient, but not necessary, to mediate PLG-Ag-induced tolerance in EAE. The durability of PLG-Ag-induced tolerance in the absence of KCs may be attributed to the distribution of PLG-Ags to LSECs, which demonstrated similar levels of PD-L1, PGE(2), and T cell stimulatory ability. Collectively, these studies provide mechanistic support for the role of liver KCs and LSECs in Ag-specific tolerance for a biomaterial platform that is currently being evaluated in clinical trials.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据