Abstract

We previously demonstrated that the formation of complexes between the DNA binding domains of the hepatocyte nuclear factor 6 (HNF6) and Forkhead Box a2 (Foxa2) transcription factors resulted in synergistic transcriptional activation of a Foxa2 target promoter. This Foxa2.HNF6 transcriptional synergy was mediated by the recruitment of CREB-binding protein (CBP) coactivator through the HNF6 Cut-Homeodomain sequences. Although the HNF6 DNA binding domain sequences are sufficient to recruit CBP coactivator for HNF6.Foxa2 transcriptional synergy, paradoxically these HNF6 Cut-Homeodomain sequences were unable to stimulate the transcription of an HNF6-dependent reporter gene. Here, we investigated whether the CBP coactivator protein played a different role in regulating HNF6 transcriptional activity. We showed that acetylation of the HNF6 protein by CBP increased both HNF6 protein stability and its ability to stimulate transcription of the glucose transporter 2 promoter. Mutation of the HNF6 Cut domain lysine 339 residue to an arginine residue abrogated CBP acetylation, which is required for HNF6 protein stability. Furthermore, the HNF6 K339R mutant protein, which failed to accumulate detected protein levels, was transcriptionally inactive and could not be stabilized by inhibiting the ubiquitin proteasome pathway. Finally, increased HNF6 protein levels stabilized the Foxa2 protein, presumably through the formation of the Foxa2.HNF6 complex. These studies show for the first time that HNF6 protein stability is controlled by CBP acetylation and provides a novel mechanism by which the activity of the CBP coactivator may regulate steady levels of two distinct liver-enriched transcription factors.

Highlights

  • Synergistic transcriptional activation of hepatocyte-specific genes required the simultaneous binding of multiple families of liver-enriched transcription factors to the DNA regulatory regions of these cell type-specific genes [1]

  • We previously demonstrated that the formation of complexes between the DNA binding domains of the hepatocyte nuclear factor 6 (HNF6) and Forkhead Box a2 (Foxa2) transcription factors resulted in synergistic transcriptional activation of a Foxa2 target promoter

  • Increased HNF6 protein levels stabilized the Foxa2 protein, presumably through the formation of the Foxa21⁄7HNF6 complex. These studies show for the first time that HNF6 protein stability is controlled by CREB-binding protein (CBP) acetylation and provides a novel mechanism by which the activity of the CBP coactivator may regulate steady levels of two distinct liver-enriched transcription factors

Read more

Summary

Introduction

Synergistic transcriptional activation of hepatocyte-specific genes required the simultaneous binding of multiple families of liver-enriched transcription factors to the DNA regulatory regions of these cell type-specific genes [1]. On the basis of homology within DNA binding domains of these regulatory hepatocyte nuclear factors (HNF), the liver-enriched transcription. Many of these liver transcription factors recruit the p300/ CREB-binding protein (CBP) family of histone acetyltransferases to activate transcription of hepatocyte-specific genes. These include the HNF4␣ [15, 16], HNF1␣ [17, 18], HNF6 [19], and C/EBP transcription factors [20]. CREB-binding protein; TTR, transthyretin; CMV, cytomegalovirus virus immediate early promoter; Glut, Ϫ188-bp glucose transporter 2; Ϫ188 Glut, Ϫ188-bp Glut; C/EBP, CCAAT/enhancer-binding protein; WT, wild type; CHx, cycloheximide; Cdk, cyclin-dependent kinase; ERK, extracellular signal-regulated kinase; E1A, adenovirus E1A protein

Methods
Results
Conclusion
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