Epinephrine regulation of the endothelial nitric-oxide synthase: roles of RAC1 and beta3-adrenergic receptors in endothelial NO signaling

J Biol Chem. 2007 Nov 9;282(45):32719-29. doi: 10.1074/jbc.M706815200. Epub 2007 Sep 12.

Abstract

beta-Adrenergic receptors (betaAR) play an important role in vasodilation, but the mechanisms whereby adrenergic pathways regulate the endothelial isoform of nitric-oxide synthase (eNOS) are incompletely understood. We found that epinephrine significantly increases eNOS activity in cultured bovine aortic endothelial cells (BAEC). Epinephrine-dependent eNOS activation was accompanied by an increase in phosphorylation of eNOS at Ser(1179) and with decreased eNOS phosphorylation at the inhibitory phosphoresidues Ser(116) and Thr(497). Epinephrine promoted activation of the small G protein Rac1 and also led to the activation of protein kinase A. All of these responses to epinephrine in BAEC were blocked by the beta(3)AR blocker SR59230A. We transfected and validated duplex small interfering RNA (siRNA) constructs to selectively "knock down" specific signaling proteins in BAEC. siRNA-mediated knockdown of Rac1 completely blocked all beta(3)AR signaling to eNOS and also abrogated epinephrine-dependent cAMP-dependent protein kinase (PKA) and Akt activation. However, siRNA-mediated knockdown of PKA did not affect Rac1 activation by epinephrine but did attenuate Akt activation by epinephrine. These findings indicate that Rac1 is an upstream regulator of beta(3)AR signaling to PKA and to eNOS and identify a novel beta(3)AR --> Rac1 --> PKA --> Akt pathway in endothelium. We exploited the p21-activated kinase pulldown assay to identify proteins associated with activated Rac1 and found that epinephrine stimulated the association of eNOS with Rac1; epinephrine-stimulated eNOS-Rac1 interactions were blocked by the beta(3)AR antagonist SR59230A. Co-transfection of eNOS cDNA with constitutively active Rac1 enhanced beta(3)AR-promoted eNOS-Rac1 association; co-transfection of eNOS with dominant negative Rac1 completely blocked the eNOS-Rac1 association. We also found that epinephrine-induced Rac1 --> PKA --> Akt pathway mediates beta(3)AR-mediated endothelial cell migration. Taken together, our data establish that the small G protein Rac1 is a key regulator of beta(3)AR signaling in cultured aortic endothelial cells with potentially important implications for the pathways involved in adrenergic modulation of eNOS pathways in the vascular wall.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Adrenergic beta-3 Receptor Antagonists
  • Adrenergic beta-Antagonists / pharmacology
  • Animals
  • Cattle
  • Cells, Cultured
  • Endothelial Cells / drug effects*
  • Endothelial Cells / metabolism*
  • Enzyme Activation / drug effects
  • Epinephrine / pharmacology*
  • Humans
  • Nitric Oxide / metabolism*
  • Nitric Oxide Synthase Type III / genetics
  • Nitric Oxide Synthase Type III / metabolism*
  • Phosphorylation / drug effects
  • Protein Binding
  • Protein Kinase Inhibitors / pharmacology
  • RNA, Small Interfering / genetics
  • Receptors, Adrenergic, beta / metabolism
  • Receptors, Adrenergic, beta-3 / metabolism*
  • Signal Transduction
  • rac1 GTP-Binding Protein / genetics
  • rac1 GTP-Binding Protein / metabolism*

Substances

  • Adrenergic beta-3 Receptor Antagonists
  • Adrenergic beta-Antagonists
  • Protein Kinase Inhibitors
  • RNA, Small Interfering
  • Receptors, Adrenergic, beta
  • Receptors, Adrenergic, beta-3
  • Nitric Oxide
  • Nitric Oxide Synthase Type III
  • rac1 GTP-Binding Protein
  • Epinephrine