Soluble CD83 modulates human-monocyte-derived macrophages toward alternative phenotype, function, and metabolism

Front Immunol. 2023 Dec 14:14:1293828. doi: 10.3389/fimmu.2023.1293828. eCollection 2023.

Abstract

Alterations in macrophage (Mφ) polarization, function, and metabolic signature can foster development of chronic diseases, such as autoimmunity or fibrotic tissue remodeling. Thus, identification of novel therapeutic agents that modulate human Mφ biology is crucial for treatment of such conditions. Herein, we demonstrate that the soluble CD83 (sCD83) protein induces pro-resolving features in human monocyte-derived Mφ biology. We show that sCD83 strikingly increases the expression of inhibitory molecules including ILT-2 (immunoglobulin-like transcript 2), ILT-4, ILT-5, and CD163, whereas activation markers, such as MHC-II and MSR-1, were significantly downregulated. This goes along with a decreased capacity to stimulate alloreactive T cells in mixed lymphocyte reaction (MLR) assays. Bulk RNA sequencing and pathway analyses revealed that sCD83 downregulates pathways associated with pro-inflammatory, classically activated Mφ (CAM) differentiation including HIF-1A, IL-6, and cytokine storm, whereas pathways related to alternative Mφ activation and liver X receptor were significantly induced. By using the LXR pathway antagonist GSK2033, we show that transcription of specific genes (e.g., PPARG, ABCA1, ABCG1, CD36) induced by sCD83 is dependent on LXR activation. In summary, we herein reveal for the first time mechanistic insights into the modulation of human Mφ biology by sCD83, which is a further crucial preclinical study for the establishment of sCD83 as a new therapeutical agent to treat inflammatory conditions.

Keywords: LXR pathway; alternative activation; checkpoint molecule; human-monocyte-derived macrophages; soluble CD83.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • CD83 Antigen*
  • Cell Differentiation
  • Humans
  • Macrophages*
  • Phenotype
  • T-Lymphocytes*

Substances

  • CD83 protein, human
  • CD83 Antigen

Grants and funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by the Deutsche Forschungsgemeinschaft (DFG) within grants SFB1181 project B03, STE432/15-1 (to ASte), by the Interdisciplinary Center of Clinical Studies (IZKF) at the University Hospital of the FAU Erlangen-Nuremberg to AS (grant A89), and the Interdisciplinary Center of Clinical Studies (IZKF) at the University Hospital of the FAU Erlangen-Nuremberg (ELAN P077) (to DR). The Else Kröner-Fresenius-Stiftung (grant 2020_EKEA.81) supported AW. The Bavarian Equal Opportunities Sponsorship funded KP-M – Realization Equal Opportunities for Women in Research and Teaching, and the ELAN Fond of the University Hospital of the FAU Erlangen-Nuremberg (grant P105). ASte was further supported by the m4 Award (grant M4-2110-0003), sponsored by the Bavarian State Ministry of Economic Affairs and Media, Energy and Technology. J-PA and GK were funded by the Interdisciplinary Center for Clinical Research of the University Hospital Erlangen (grant J91).