Simultaneous deletion of ORMDL1 and ORMDL3 proteins disrupts immune cell homeostasis

Front Immunol. 2024 Apr 23:15:1376629. doi: 10.3389/fimmu.2024.1376629. eCollection 2024.

Abstract

ORMDL3 is a prominent member of a family of highly conserved endoplasmic reticulum resident proteins, ORMs (ORM1 and ORM2) in yeast, dORMDL in Drosophila and ORMDLs (ORMDL1, ORMDL2, and ORMDL3) in mammals. ORMDL3 mediates feedback inhibition of de novo sphingolipid synthesis. Expression levels of ORMDL3 are associated with the development of inflammatory and autoimmune diseases including asthma, systemic lupus erythematosus, type 1 diabetes mellitus and others. It has been shown that simultaneous deletions of other ORMDL family members could potentiate ORMDL3-induced phenotypes. To understand the complex function of ORMDL proteins in immunity in vivo, we analyzed mice with single or double deletions of Ormdl genes. In contrast to other single and double knockouts, simultaneous deletion of ORMDL1 and ORMDL3 proteins disrupted blood homeostasis and reduced immune cell content in peripheral blood and spleens of mice. The reduced number of splenocytes was not caused by aberrant immune cell homing. A competitive bone marrow transplantation assay showed that the development of Ormdl1-/-/Ormdl3-/- B cells was dependent on lymphocyte intrinsic factors. Highly increased sphingolipid production was observed in the spleens and bone marrow of Ormdl1-/-/Ormdl3-/- mice. Slight, yet significant, increase in some sphingolipid species was also observed in the spleens of Ormdl3-/- mice and in the bone marrow of both, Ormdl1-/- and Ormdl3-/- single knockout mice. Taken together, our results demonstrate that the physiological expression of ORMDL proteins is critical for the proper development and circulation of lymphocytes. We also show cell-type specific roles of individual ORMDL family members in the production of different sphingolipid species.

Keywords: B cells; ORMDL proteins; lymphocyte; sphingolipids; spleen.

Publication types

  • Research Support, Non-U.S. Gov't
  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • B-Lymphocytes / immunology
  • B-Lymphocytes / metabolism
  • Gene Deletion
  • Homeostasis*
  • Membrane Proteins* / genetics
  • Membrane Proteins* / metabolism
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout*
  • Sphingolipids / metabolism
  • Spleen / immunology
  • Spleen / metabolism

Substances

  • Membrane Proteins
  • ORMDL3 protein, mouse
  • Sphingolipids

Grants and funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by projects 20-16481S, 23-07736S and 24-12572S from the Czech Science Foundation and by the Institute of Molecular Genetics supported by the Czech Academy of Sciences RVO 68378050. LK acknowledges the support of the institutional programs of Charles University in Prague (UNCE/MED/007 and Cooperation/DIAG/1FM). LD was supported in part by the First Faculty of Medicine, Charles University, Prague (Student number 85213635). The study was partially supported by the National Institute for Cancer Research (Program EXCELES, ID Project No. LX22NPO5102)—Funded by the European Union—Next Generation EU.