The lysine deacetylase Sirtuin 1 modulates the localization and function of the Notch1 receptor in regulatory T cells

N Marcel, LR Perumalsamy, SK Shukla, A Sarin - Science signaling, 2017 - science.org
Science signaling, 2017science.org
The ability to tune cellular functions in response to nutrient availability has important
consequences for immune homeostasis. The activity of the receptor Notch in regulatory T
(Treg) cells, which suppress the functions of effector T cells, is indispensable for Treg cell
survival under conditions of diminished nutrient supply. Anti-apoptotic signaling induced by
the Notch1 intracellular domain (NIC) originates from the cytoplasm and is spatially
decoupled from the nuclear, largely transcriptional functions of NIC. We showed that Sirtuin …
The ability to tune cellular functions in response to nutrient availability has important consequences for immune homeostasis. The activity of the receptor Notch in regulatory T (Treg) cells, which suppress the functions of effector T cells, is indispensable for Treg cell survival under conditions of diminished nutrient supply. Anti-apoptotic signaling induced by the Notch1 intracellular domain (NIC) originates from the cytoplasm and is spatially decoupled from the nuclear, largely transcriptional functions of NIC. We showed that Sirtuin 1 (Sirt1), which is an NAD+ (nicotinamide adenine dinucleotide)–dependent lysine deacetylase that inhibits NIC-dependent gene transcription, stabilized NIC proximal to the plasma membrane to promote the survival and function of activated Treg cells. Sirt1 was required for NIC-dependent protection from apoptosis in cell lines but not for the activity of the anti-apoptotic protein Bcl-xL. In addition, a variant NIC protein in which four lysines were mutated to arginines (NIC4KR) retained anti-apoptotic activity, but was not regulated by Sirt1, and reconstituted the functions of nonnuclear NIC in Notch1-deficient Treg cells. Loss of Sirt1 compromised Treg cell survival, resulting in antigen-induced T cell proliferation and inflammation in two mouse models. Thus, the Sirt1-Notch interaction may constitute an important checkpoint that tunes noncanonical Notch1 signaling.
AAAS