Abstract

In skeletal muscle, the Rab GTPase-activating (GAP) protein TBC1D1 is phosphorylated by AKT and AMP-activated protein kinase (AMPK) in response to insulin and muscle contraction. Genetic ablation of Tbc1d1 or mutation of distinct phosphorylation sites impairs intracellular GLUT4 retention and GLUT4 traffic, presumably through alterations of the activation state of downstream Rab GTPases. Previous studies have focused on characterizing the C-terminal GAP domain of TBC1D1 that lacks the known phosphorylation sites, as well as putative regulatory domains. As a result, it has been unclear how phosphorylation of TBC1D1 would regulate its activity. In the present study, we have expressed, purified, and characterized recombinant full-length TBC1D1 in Sf9 insect cells via the baculovirus system. Full-length TBC1D1 showed RabGAP activity toward GLUT4-associated Rab8a, Rab10, and Rab14, indicating similar substrate specificity as the truncated GAP domain. However, the catalytic activity of the full-length TBC1D1 was markedly higher than that of the GAP domain. Although in vitro phosphorylation of TBC1D1 by AKT or AMPK increased 14-3-3 binding, it did not alter the intrinsic RabGAP activity. However, we found that TBC1D1 interacts through its N-terminal PTB domains with the cytoplasmic domain of the insulin-regulated aminopeptidase, a resident protein of GLUT4 storage vesicles, and this binding is disrupted by phosphorylation of TBC1D1 by AKT or AMPK. In summary, our findings suggest that other regions outside the GAP domain may contribute to the catalytic activity of TBC1D1. Moreover, our data indicate that recruitment of TBC1D1 to GLUT4-containing vesicles and not its GAP activity is regulated by insulin and contraction-mediated phosphorylation.

Highlights

  • TBC1D1 and TBC1D4 (Tre-2/Bub2/Cdc16 domain family member 1 and 4)2 are Rab-GTPase-activating proteins (RabGAPs) involved in insulin signaling, GLUT4 translocation [1,2,3], and may play roles in obesity and type 2 diabetes (4 –6)

  • Insulin- and contraction-mediated Ser/Thr phosphorylation of the ϳ150 –160-kDa RabGAPs TBC1D1 and TBC1D4 by AKT and AMPK has been associated with alterations in the activation state of downstream Rab GTPases, suggesting that the GAP activity might be directly or indirectly regulated by phosphorylation [7, 17, 29]

  • Samples were separated by SDS-PAGE and co-precipitated FLAG-TBC1D1 was analyzed by Western blotting

Read more

Summary

Results

We generated a baculovirus coding for the long isoform of the murine TBC1D1 that is predominantly expressed in skeletal muscle [4]. Purified GST-Rab was loaded with [␥-32P]GTP and incubated either alone, or with full-length TBC1D1, or the GAP mutant (R854K) of TBC1D1 as described under “Experimental procedures.”. Compared with the truncated GAP domain, the full-length protein displayed markedly higher activity toward Rab as substrate (Fig. 2B). We investigated the direct impact of AKT2 and AMPK-mediated phosphorylation on the GAP activity of TBC1D1 using Rab as substrate. IMAC purified TBC1D1 was phosphorylated by either AKT2 or AMPK for 30 min, incubated with purified, [␥-32P]GTP-loaded Rab and the amount of released [32P]phosphate was determined as described under “Experimental procedures.”. To test whether phosphorylation-induced 14-3-3 binding alters the GAP activity, TBC1D1 was phosphorylated by AKT2 or AMPK, and association of GST–14-3-3 was determined by GST-pulldown assay as described under “Experimental procedures.”.

Discussion
DNA constructs
Protein expression and purification
Size exclusion chromatography
Mass spectrometry
Kinase assays
GST pulldown reactions
RabGAP assay
Data analysis
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call