A mitotic GlcNAcylation/phosphorylation signaling complex alters the posttranslational state of the cytoskeletal protein vimentin

C Slawson, T Lakshmanan, S Knapp… - Molecular biology of the …, 2008 - Am Soc Cell Biol
C Slawson, T Lakshmanan, S Knapp, GW Hart
Molecular biology of the cell, 2008Am Soc Cell Biol
O-linked β-N-acetylglucosamine (O-GlcNAc) is a highly dynamic intracellular protein
modification responsive to stress, hormones, nutrients, and cell cycle stage. Alterations in O-
GlcNAc addition or removal (cycling) impair cell cycle progression and cytokinesis, but the
mechanisms are not well understood. Here, we demonstrate that the enzymes responsible
for O-GlcNAc cycling, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) are in a
transient complex at M phase with the mitotic kinase Aurora B and protein phosphatase 1 …
O-linked β-N-acetylglucosamine (O-GlcNAc) is a highly dynamic intracellular protein modification responsive to stress, hormones, nutrients, and cell cycle stage. Alterations in O-GlcNAc addition or removal (cycling) impair cell cycle progression and cytokinesis, but the mechanisms are not well understood. Here, we demonstrate that the enzymes responsible for O-GlcNAc cycling, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) are in a transient complex at M phase with the mitotic kinase Aurora B and protein phosphatase 1. OGT colocalized to the midbody during telophase with Aurora B. Furthermore, these proteins coprecipitated with each other in a late mitotic extract. The complex was stable under Aurora inhibition; however, the total cellular levels of O-GlcNAc were increased and the localization of OGT was decreased at the midbody after Aurora inhibition. Vimentin, an intermediate filament protein, is an M phase substrate for both Aurora B and OGT. Overexpression of OGT or OGA led to defects in mitotic phosphorylation on multiple sites, whereas OGT overexpression increased mitotic GlcNAcylation of vimentin. OGA inhibition caused a decrease in vimentin late mitotic phosphorylation but increased GlcNAcylation. Together, these data demonstrate that the O-GlcNAc cycling enzymes associate with kinases and phosphatases at M phase to regulate the posttranslational status of vimentin.
Am Soc Cell Biol