KMT2D regulates activation, localization, and integrin expression by T-cells

Front Immunol. 2024 May 3:15:1341745. doi: 10.3389/fimmu.2024.1341745. eCollection 2024.

Abstract

Individuals with Kabuki syndrome present with immunodeficiency; however, how pathogenic variants in the gene encoding the histone-modifying enzyme lysine methyltransferase 2D (KMT2D) lead to immune alterations remain poorly understood. Following up on our prior report of KMT2D-altered integrin expression in B-cells, we performed targeted analyses of KMT2D's influence on integrin expression in T-cells throughout development (thymocytes through peripheral T-cells) in murine cells with constitutive- and conditional-targeted Kmt2d deletion. Using high-throughput RNA-sequencing and flow cytometry, we reveal decreased expression (both at the transcriptional and translational levels) of a cluster of leukocyte-specific integrins, which perturb aspects of T-cell activation, maturation, adhesion/localization, and effector function. H3K4me3 ChIP-PCR suggests that these evolutionary similar integrins are under direct control of KMT2D. KMT2D loss also alters multiple downstream programming/signaling pathways, including integrin-based localization, which can influence T-cell populations. We further demonstrated that KMT2D deficiency is associated with the accumulation of murine CD8+ single-positive (SP) thymocytes and shifts in both human and murine peripheral T-cell populations, including the reduction of the CD4+ recent thymic emigrant (RTE) population. Together, these data show that the targeted loss of Kmt2d in the T-cell lineage recapitulates several distinct features of Kabuki syndrome-associated immune deficiency and implicates epigenetic mechanisms in the regulation of integrin signaling.

Keywords: Itgal; Itgb7; KS1-associated immune deficiency (KSAID); Kabuki syndrome (KS); integrin switching; recent thymic emigrant (RTE); thymocyte.

MeSH terms

  • Abnormalities, Multiple
  • Animals
  • DNA-Binding Proteins / genetics
  • DNA-Binding Proteins / metabolism
  • Face / abnormalities
  • Gene Expression Regulation
  • Hematologic Diseases
  • Histone-Lysine N-Methyltransferase / genetics
  • Histone-Lysine N-Methyltransferase / metabolism
  • Humans
  • Integrins* / genetics
  • Integrins* / metabolism
  • Lymphocyte Activation* / genetics
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Myeloid-Lymphoid Leukemia Protein
  • Neoplasm Proteins / genetics
  • Neoplasm Proteins / immunology
  • Neoplasm Proteins / metabolism
  • Signal Transduction
  • T-Lymphocytes / immunology
  • T-Lymphocytes / metabolism
  • Vestibular Diseases / genetics
  • Vestibular Diseases / immunology
  • Vestibular Diseases / metabolism

Substances

  • Kmt2d protein, mouse

Supplementary concepts

  • Kabuki syndrome

Grants and funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. HB and LZ were funded by the Louma G. Foundation, which specifically supported this work. HB is also funded by The Icelandic Research Fund (#217988, #195835, and #206806) and the Icelandic Technology Development Fund (#2010588-0611). The work of SP, AL, and AB was funded by a Center for Pediatric Genomics (CpG) grant from the Cincinnati Children’s Research Foundation and supported by the Center for Stem Cell and Organoid Medicine (CuSTOM) at Cincinnati Children’s Hospital Medical Center. Flow cytometric data generated by SP were acquired using equipment maintained by the Research Flow Cytometry Core in the Division of Rheumatology at Cincinnati Children’s Hospital Medical Center (CCHMC). Work was supported in part by the Center for Rheumatic Disease Research (NIH AR070549) and Digestive Health Center (NIDDK P30 DK078392). Content was placed on BioRxiv: The preprint server for biology on October 07, 2022 as doi: https://doi.org/10.1101/2022.10.04.510662 (90).