Autoantibodies from patients with kidney allograft vasculopathy stimulate a proinflammatory switch in endothelial cells and monocytes mediated via GPCR-directed PAR1-TNF-α signaling

Front Immunol. 2023 Oct 30:14:1289744. doi: 10.3389/fimmu.2023.1289744. eCollection 2023.

Abstract

Non-HLA-directed regulatory autoantibodies (RABs) are known to target G-protein coupled receptors (GPCRs) and thereby contribute to kidney transplant vasculopathy and failure. However, the detailed underlying signaling mechanisms in human microvascular endothelial cells (HMECs) and immune cells need to be clarified in more detail. In this study, we compared the immune stimulatory effects and concomitant intracellular and extracellular signaling mechanisms of immunoglobulin G (IgG)-fractions from kidney transplant patients with allograft vasculopathy (KTx-IgG), to that from patients without vasculopathy, or matched healthy controls (Con-IgG). We found that KTx-IgG from patients with vasculopathy, but not KTx-IgG from patients without vasculopathy or Con-IgG, elicits HMEC activation and subsequent upregulation and secretion of tumor necrosis factor alpha (TNF-α) from HMECs, which was amplified in the presence of the protease-activated thrombin receptor 1 (PAR1) activator thrombin, but could be omitted by selectively blocking the PAR1 receptor. The amount and activity of the TNF-α secreted by HMECs stimulated with KTx-IgG from patients with vasculopathy was sufficient to induce subsequent THP-1 monocytic cell activation. Furthermore, AP-1/c-FOS, was identified as crucial transcription factor complex controlling the KTx-IgG-induced endothelial TNF-α synthesis, and mircoRNA-let-7f-5p as a regulatory element in modulating the underlying signaling cascade. In conclusion, exposure of HMECs to KTx-IgG from patients with allograft vasculopathy, but not KTx-IgG from patients without vasculopathy or healthy Con-IgG, triggers signaling through the PAR1-AP-1/c-FOS-miRNA-let7-axis, to control TNF-α gene transcription and TNF-α-induced monocyte activation. These observations offer a greater mechanistic understanding of endothelial cells and subsequent immune cell activation in the clinical setting of transplant vasculopathy that can eventually lead to transplant failure, irrespective of alloantigen-directed responses.

Keywords: autoantibodies; chronic kidney disease (CKD); end-stage renal disease (ESRD); endothelial cells (ECs); kidney allograft vasculopathy; kidney transplantation (KTx); non-HLA-directed regulatory autoantibodies (RABs); tumor necrosis factor-alpha (TNF-α).

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Allografts
  • Autoantibodies
  • Endothelial Cells / physiology
  • Humans
  • Immunoglobulin G
  • Kidney
  • Kidney Diseases*
  • Monocytes
  • Receptor, PAR-1
  • Thrombin*
  • Transcription Factor AP-1
  • Tumor Necrosis Factor-alpha / metabolism

Substances

  • Autoantibodies
  • Immunoglobulin G
  • Receptor, PAR-1
  • Thrombin
  • Transcription Factor AP-1
  • Tumor Necrosis Factor-alpha

Grants and funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. GM’s and RC’s contributions were made possible by funding from the German Federal Ministry for Education and Research (BMBF) and German Research Foundation (DFG; projects Nephroprotection #394046635, subproject A03, as part of CRC 1365, and EXPAND-PD; CA2816/1-1) and through the BIH Center for Regenerative Therapies (BCRT) and Berlin-Brandenburg School for Regenerative Therapies (BSRT, GSC203), respectively, and in part by the European Union’s Horizon 2020 Research and Innovation Program under grant agreements No 733006 (PACE) and 779293 (HIPGEN) and 754995 (EU-TRAIN). OC-M’s contributions were made possible by The São Paulo Research Foundation (FAPESP 2018/18886-9, 2020/01688-0, and 2020/07069-0). We acknowledge financial support from the Open Access Publication Fund of Charité Universitätsmedizin Berlin and the DFG.