Abstract

The G protein-coupled ghrelin receptor GHSR1a is a potential pharmacological target for treating obesity and addiction because of the critical role ghrelin plays in energy homeostasis and dopamine-dependent reward. GHSR1a enhances growth hormone release, appetite, and dopamine signaling through G(q/11), G(i/o), and G(12/13) as well as β-arrestin-based scaffolds. However, the contribution of individual G protein and β-arrestin pathways to the diverse physiological responses mediated by ghrelin remains unknown. To characterize whether a signaling bias occurs for GHSR1a, we investigated ghrelin signaling in a number of cell-based assays, including Ca(2+) mobilization, serum response factor response element, stress fiber formation, ERK1/2 phosphorylation, and β-arrestin translocation, utilizing intracellular second loop and C-tail mutants of GHSR1a. We observed that GHSR1a and β-arrestin rapidly form metastable plasma membrane complexes following exposure to an agonist, but replacement of the GHSR1a C-tail by the tail of the vasopressin 2 receptor greatly stabilizes them, producing complexes observable on the plasma membrane and also in endocytic vesicles. Mutations of the contiguous conserved amino acids Pro-148 and Leu-149 in the GHSR1a intracellular second loop generate receptors with a strong bias to G protein and β-arrestin, respectively, supporting a role for conformation-dependent signaling bias in the wild-type receptor. Our results demonstrate more balance in GHSR1a-mediated ERK signaling from G proteins and β-arrestin but uncover an important role for β-arrestin in RhoA activation and stress fiber formation. These findings suggest an avenue for modulating drug abuse-associated changes in synaptic plasticity via GHSR1a and indicate the development of GHSR1a-biased ligands as a promising strategy for selectively targeting downstream signaling events.

Highlights

  • The G protein coupled receptor GHSR1a mediates feeding and addictive behaviors

  • ␤-Arrestin-2 Interaction with the GHSR1a in HEK293 Cells— A specific cluster of phosphorylated serine/threonine residues in the C terminus of class B G protein-coupled receptor (GPCR) enables them to form stable complexes with ␤-arrestin during endocytosis [37]. These complexes are readily apparent by confocal microscopy because they distinctly label the membranes of early endosomes but are absent from their interior [38, 39], and we previously identified the location of these clusters for representative GPCRs by C-tail mutagenesis [37, 39]

  • L585 was more potent than ghrelin in stimulating Ca2ϩ, with EC50 values of 2.7 Ϯ 1.2 nM and 93.5 Ϯ 11.6 nM, respectively (Fig. 4A). This order of potency is consistent with reports on similar small molecule agonists of the ghrelin receptor [4], but it is the reverse of what we found for the ␤-arrestin translocation assay

Read more

Summary

Background

The G protein coupled receptor GHSR1a mediates feeding and addictive behaviors. Results: Mutagenesis of the second intracellular loop of GHSR1a generates biased receptors, favoring distinct signaling events. Our results demonstrate more balance in GHSR1a-mediated ERK signaling from G proteins and ␤-arrestin but uncover an important role for ␤-arrestin in RhoA activation and stress fiber formation These findings suggest an avenue for modulating drug abuse-associated changes in synaptic plasticity via GHSR1a and indicate the development of GHSR1a-biased ligands as a promising strategy for selectively targeting downstream signaling events. We report that GHSR1a forms transient complexes with ␤-arrestin-2 on the cell membrane that can be stabilized by the addition of phosphate acceptor sites in the receptor tail or destabilized by substitution of a conserved proline residue in the receptor ICL2 This interaction with ␤-arrestin is essential for the GHSR1a-mediated induction of RhoA signaling and actin remodeling, whereas GHSR1a-mediated ERK1/2 phosphorylation involves both ␤-arrestin and G protein components. Most significantly, mutating amino acid residues for the interaction with ␤-arrestin or G proteins in ICL2 generated signaling-biased receptors, potentially allowing future studies to determine their individual contribution to various in vivo physiological processes mediated by ghrelin

EXPERIMENTAL PROCEDURES
RESULTS
Findings
DISCUSSION
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