Abstract

The transcriptional activity of the glucocorticoid receptor (GR) is co-determined by its ability to recruit a vast and varying number of cofactors. We here identify Striatin-3 (STRN3) as a novel interaction partner of GR that interferes with GR’s ligand-dependent transactivation capacity. Remarkably, STRN3 selectively affects only GR-dependent transactivation and leaves GR-dependent transrepression mechanisms unhampered. We found that STRN3 down-regulates GR transactivation by an additional recruitment of the catalytic subunit of protein phosphatase 2A (PPP2CA) to GR. We hypothesize the existence of a functional trimeric complex in the nucleus, able to dephosphorylate GR at serine 211, a known marker for GR transactivation in a target gene-dependent manner. The presence of STRN3 appears an absolute prerequisite for PPP2CA to engage in a complex with GR. Herein, the C-terminal domain of GR is essential, reflecting ligand-dependency, yet other receptor parts are also needed to create additional contacts with STRN3.

Highlights

  • Glucocorticoids (GC) exert effects at multiple levels of cellular functionality, including energy metabolism, cell fate and immune response

  • Since ECI2 is a PPARα/ GRα co-controlled gene[34] and PPARα may be a target of STRN3 (Fig. 4B) it is tempting to speculate that the different regulation of ECI2 might be ascribed to an increased level control of this particular gene promoter

  • By using a glucocorticoid receptor (GR) mutant of serine 211 (S211) (S211A) we showed that the negative impact of STRN3 on GR transactivation is phosphorylation-dependent

Read more

Summary

Introduction

Glucocorticoids (GC) exert effects at multiple levels of cellular functionality, including energy metabolism, cell fate and immune response. GR-mediated gene promoter activation involves DNA binding of a homodimeric GR on a palindromic GC response element (GRE) in the promoter (simple GRE), from a coordinated DNA binding of a GR/transcription factor complex onto a so-called composite GRE or from a GR/transcription factor tethering mechanism. The latter two mechanisms can form the basis for GR-mediated promoter repression[5, 6]. Exogenous GC treatment induces GR dimer assembly to classic palindromic site-containing promoters of ligand-dependent genes. This event is at the expense of monomeric GR binding which vanishes from promoters of repressed genes[8]. Our data support a role for STRN3 as an important check-point for GR functionality

Methods
Results
Conclusion
Full Text
Paper version not known

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