Role of Brg1 and HDAC2 in GR trans-repression of the pituitary POMC gene and misexpression in Cushing disease

  1. Steve Bilodeau1,
  2. Sophie Vallette-Kasic1,
  3. Yves Gauthier1,
  4. Dominique Figarella-Branger2,
  5. Thierry Brue2,
  6. France Berthelet3,
  7. André Lacroix3,
  8. Dalia Batista4,
  9. Constantine Stratakis4,
  10. Jeanette Hanson5,
  11. Björn Meij5, and
  12. Jacques Drouin1,6
  1. 1Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada;
  2. 2Laboratoire Interactions cellulaires neuroendocriniennes (ICNE), Université de la Méditerranée, Centre national de la recherche scientifique (CNRS) UMR 6544, Institut Jean-Roche, 13385 Marseille, France;
  3. 3Department of Medicine, Research Center, Hotel-Dieu du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec H2W 1T8, Canada;
  4. 4Section on Endocrinology and Genetics (SEGEN), Developmental Endocrinology Branch (DEB), National Institute of Child Health and Human Development (NICHD), Bethesda, Maryland 20892, USA;
  5. 5Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, NL-3508-TD Utrecht, Netherlands

Abstract

Negative feedback regulation of the proopiomelanocortin (POMC) gene by the glucocorticoid (Gc) receptor (GR) is a critical feature of the hypothalamo–pituitary–adrenal axis, and it is in part exerted by trans-repression between GR and the orphan nuclear receptors related to NGFI-B. We now show that Brg1, the ATPase subunit of the Swi/Snf complex, is essential for this trans-repression and that Brg1 is required in vivo to stabilize interactions between GR and NGFI-B as well as between GR and HDAC2. Whereas Brg1 is constitutively present at the POMC promoter, recruitment of GR and HDAC2 is ligand-dependent and results in histone H4 deacetylation of the POMC locus. In addition, GR-dependent repression inhibits promoter clearance by RNA polymerase II. Thus, corecruitment of repressor and activator at the promoter and chromatin modification jointly contribute to trans-repression initiated by direct interactions between GR and NGFI-B. Loss of Brg1 or HDAC2 should therefore produce Gc resistance, and we show that ∼50% of Gc-resistant human and dog corticotroph adenomas, which are the hallmark of Cushing disease, are deficient in nuclear expression of either protein. In addition to providing a molecular basis for Gc resistance, these deficiencies may also contribute to the tumorigenic process.

Keywords

Footnotes

| Table of Contents

Life Science Alliance