PRMT5 regulates epigenetic changes in suppressive Th1-like iTregs in response to IL-12 treatment

Front Immunol. 2024 Jan 8:14:1292049. doi: 10.3389/fimmu.2023.1292049. eCollection 2023.

Abstract

Background: Induced regulatory T cells (iTregs) are a heterogeneous population of immunosuppressive T cells with therapeutic potential. Treg cells show a range of plasticity and can acquire T effector-like capacities, as is the case for T helper 1 (Th1)-like iTregs. Thus, it is important to distinguish between functional plasticity and lineage instability. Aplastic anemia (AA) is an autoimmune disorder characterized by immune-mediated destruction of hematopoietic stem and progenitor cells in the bone marrow (BM). Th1-like 1 iTregs can be potent suppressors of aberrant Th1-mediated immune responses such as those that drive AA disease progression. Here we investigated the function of the epigenetic enzyme, protein arginine methyltransferase 5 (PRMT5), its regulation of the iTreg-destabilizing deacetylase, sirtuin 1 (Sirt1) in suppressive Th1-like iTregs, and the potential for administering Th1-like iTregs as a cell-based therapy for AA.

Methods: We generated Th1-like iTregs by culturing iTregs with IL-12, then assessed their suppressive capacity, expression of iTreg suppression markers, and enzymatic activity of PRMT5 using histone symmetric arginine di-methylation (H3R2me2s) as a read out. We used ChIP sequencing on Th1 cells, iTregs, and Th1-like iTregs to identify H3R2me2s-bound genes unique to Th1-like iTregs, then validated targets using CHiP-qPCR. We knocked down PRMT5 to validate its contribution to Th1-like iTreg lineage commitment. Finally we tested the therapeutic potential of Th1-like iTregs using a Th1-mediated mouse model of AA.

Results: Exposing iTregs to the Th1 cytokine, interleukin-12 (IL-12), during early events of differentiation conveyed increased suppressive function. We observed increased PRMT5 enzymatic activity, as measured by H3R2me2s, in Th1-like iTregs, which was downregulated in iTregs. Using ChIP-sequencing we discovered that H3R2me2s is abundantly bound to the Sirt1 promoter region in Th1-like iTregs to negatively regulate its expression. Furthermore, administering Th1-like iTregs to AA mice provided a survival benefit.

Conclusions: Knocking down PRMT5 in Th1-like iTregs concomitantly reduced their suppressive capacity, supporting the notion that PRMT5 is important for the superior suppressive capacity and stability of Th1-like iTregs. Conclusively, therapeutic administration of Th1-like iTregs in a mouse model of AA significantly extended their survival and they may have therapeutic potential.

Keywords: T helper type 1-like induced regulatory T cells (Th1-like iTregs); aplastic anemia (AA); bone marrow failure (BMF); interleukin-12 (IL-12); post-translational modifications (PTMs); protein arginine methyltransferase 5 (PRMT5); sirtuin 1 (SIRT1); symmetric histone di-methylation (H3R2me2s).

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Anemia, Aplastic*
  • Animals
  • Cell Differentiation / genetics
  • Cytokines
  • Disease Models, Animal
  • Epigenesis, Genetic*
  • Interleukin-12* / pharmacology
  • Mice
  • Protein-Arginine N-Methyltransferases* / genetics
  • Sirtuin 1

Substances

  • Cytokines
  • Interleukin-12
  • Sirtuin 1
  • Prmt5 protein, mouse
  • Protein-Arginine N-Methyltransferases

Grants and funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. NJ was supported in part by DoD W81XWH2010536 (GT).