Frontiers in Endocrinology (May 2019)

O-GlcNacylation Links TxNIP to Inflammasome Activation in Pancreatic β Cells

  • Gaelle Filhoulaud,
  • Gaelle Filhoulaud,
  • Gaelle Filhoulaud,
  • Fadila Benhamed,
  • Fadila Benhamed,
  • Fadila Benhamed,
  • Patrick Pagesy,
  • Patrick Pagesy,
  • Patrick Pagesy,
  • Caroline Bonner,
  • Caroline Bonner,
  • Yann Fardini,
  • Yann Fardini,
  • Yann Fardini,
  • Anissa Ilias,
  • Anissa Ilias,
  • Jamileh Movassat,
  • Jamileh Movassat,
  • Anne-Françoise Burnol,
  • Anne-Françoise Burnol,
  • Anne-Françoise Burnol,
  • Sandra Guilmeau,
  • Sandra Guilmeau,
  • Sandra Guilmeau,
  • Julie Kerr-Conte,
  • François Pattou,
  • François Pattou,
  • Tarik Issad,
  • Tarik Issad,
  • Tarik Issad,
  • Catherine Postic,
  • Catherine Postic,
  • Catherine Postic

DOI
https://doi.org/10.3389/fendo.2019.00291
Journal volume & issue
Vol. 10

Abstract

Read online

Thioredoxin interacting protein (TxNIP), which strongly responds to glucose, has emerged as a central mediator of glucotoxicity in pancreatic β cells. TxNIP is a scaffold protein interacting with target proteins to inhibit or stimulate their activity. Recent studies reported that high glucose stimulates the interaction of TxNIP with the inflammasome protein NLRP3 (NLR family, pyrin domain containing 3) to increase interleukin-1 β (IL1β) secretion by pancreatic β cells. To better understand the regulation of TxNIP by glucose in pancreatic β cells, we investigated the implication of O-linked β-N-acetylglucosamine (O-GlcNAcylation) in regulating TxNIP at the posttranslational level. O-GlcNAcylation of proteins is controlled by two enzymes: the O-GlcNAc transferase (OGT), which transfers a monosaccharide to serine/threonine residues on target proteins, and the O-GlcNAcase (OGA), which removes it. Our study shows that TxNIP is subjected to O-GlcNAcylation in response to high glucose concentrations in β cell lines. Modification of the O-GlcNAcylation pathway through manipulation of OGT or OGA expression or activity significantly modulates TxNIP O-GlcNAcylation in INS1 832/13 cells. Interestingly, expression and O-GlcNAcylation of TxNIP appeared to be increased in islets of diabetic rodents. At the mechanistic level, the induction of the O-GlcNAcylation pathway in human and rat islets promotes inflammasome activation as evidenced by enhanced cleaved IL1β. Overexpression of OGT in HEK293 or INS1 832/13 cells stimulates TxNIP and NLRP3 interaction, while reducing TxNIP O-GlcNAcylation through OGA overexpression destabilizes this interaction. Altogether, our study reveals that O-GlcNAcylation represents an important regulatory mechanism for TxNIP activity in β cells.

Keywords