Your browser doesn't support javascript.
loading
Selenoprotein I deficiency in T cells promotes differentiation into tolerant phenotypes while decreasing Th17 pathology.
Ma, Chi; Hoffmann, FuKun W; Nunes, Lance G; Urena, Frank; Andrukhiv, Anastasia; Gerschenson, Mariana; Pitts, Matthew W; Hoffmann, Peter R.
Afiliação
  • Ma C; Department of Cell and Molecular Bi ology, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA.
  • Hoffmann FW; Department of Cell and Molecular Bi ology, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA.
  • Nunes LG; Department of Cell and Molecular Bi ology, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA.
  • Urena F; Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA.
  • Andrukhiv A; Department of Cell and Molecular Bi ology, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA.
  • Gerschenson M; Department of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, USA.
  • Pitts MW; Department of Cell and Molecular Bi ology, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA.
  • Hoffmann PR; Department of Cell and Molecular Bi ology, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA.
J Leukoc Biol ; 112(6): 1387-1397, 2022 12.
Article em En | MEDLINE | ID: mdl-35916034
ABSTRACT
Selenoprotein I (SELENOI) is an ethanolamine phospholipid transferase contributing to cellular metabolism and the synthesis of glycosylphosphatidylinositol (GPI) anchors. SELENOI knockout (KO) in T cells has been shown to impair metabolic reprogramming during T cell activation and reduce GPI-anchored Thy-1 levels, which are both crucial for Th17 differentiation. This suggests SELENOI may be important for Th17 differentiation, and we found that SELENOI was indeed up-regulated early during the activation of naïve CD4+ T cells in Th17 conditions. SELENOI KO reduced RORγt mRNA levels by decreasing SOX5 and STAT3 binding to promoter and enhancer regions in the RORC gene encoding this master regulator of Th17 cell differentiation. Differentiation of naïve CD4+ T cells into inflammatory versus tolerogenic Th cell subsets was analyzed and results showed that SELENOI deficiency skewed differentiation away from pathogenic Th17 cells (RORγt+ and IL-17A+ ) while promoting tolerogenic phenotypes (Foxp3+ and IL-10+ ). Wild-type and T cell-specific SELENOI KO mice were subjected to experimental autoimmune encephalitis (EAE), with KO mice exhibiting diminished clinical symptoms, reduced CNS pathology and decreased T cell infiltration. Flow cytometry showed that SELENOI T cell KO mice exhibited lower CD4+ RORγt+ and CD4+ IL-17A+ T cells and higher CD4+ CD25+ FoxP3+ T cells in CNS tissues of mice subjected to EAE. Thus, the metabolic enzyme SELENOI is up-regulated to promote RORγt transcription that drives Th17 differentiation, and SELENOI deficiency shifts differentiation toward tolerogenic phenotypes while protecting against pathogenic Th17 responses.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares / Células Th17 Limite: Animals Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares / Células Th17 Limite: Animals Idioma: En Ano de publicação: 2022 Tipo de documento: Article