Abstract

We have previously shown that the mammalian gonadotropin-releasing hormone receptor (GnRHR), a unique G-protein-coupled receptor (GPCR) lacking an intracellular carboxyl tail (C-tail), does not follow a beta-arrestin-dependent internalization pathway. However, internalization of a chimeric GnRHR with the thyrotropin-releasing hormone receptor (TRHR) C-tail does utilize beta-arrestin. Here, we have investigated the sites within the intracellular C-tail domain that are important for conferring beta-arrestin-dependent internalization. In contrast to the chimeric GnRHR with a TRHR C-tail, a chimeric GnRHR with the catfish GnRHR C-tail is not beta-arrestin-dependent. Sequence comparisons between these chimeric receptors show three consensus phosphorylation sites for casein kinase II (CKII) in the TRHR C-tail but none in the catfish GnRHR C-tail. We thus investigated a role for CKII sites in determining GPCR internalization via beta-arrestin. Sequential introduction of three CKII sites into the chimera with the catfish C-tail (H354D,A366E,G371D) resulted in a change in the pattern of receptor phosphorylation and beta-arrestin-dependence, which only occurred when all three sites were introduced. Conversely, mutation of the putative CKII sites (T365A,T371A,S383A) in the C-tail of a beta-arrestin-sensitive GPCR, the TRHR, resulted in decreased receptor phosphorylation and a loss of beta-arrestin-dependence. Mutation of all three CKII sites was necessary before a loss of beta-arrestin-dependence was observed. Visualization of beta-arrestin/GFP redistribution confirmed a loss or gain of beta-arrestin sensitivity for receptor mutants. Internalization of receptors without C-tail CKII sites was promoted by a phosphorylation-independent beta-arrestin mutant (R169E), suggesting that these receptors do not contain the necessary phosphorylation sites required for beta-arrestin-dependent internalization. Apigenin, a specific CKII inhibitor, blocked the increase in receptor internalization by beta-arrestin, thus providing further support for the involvement of CKII. This study presents evidence of a novel role for C-tail CKII consensus sites in targeting these GPCRs to the beta-arrestin-dependent pathway.

Highlights

  • §§ Supported in part by a Block grant from the National Health and Medical Research Council of Australia to the Baker Medical Research Institute

  • We have previously shown that the mammalian gonadotropin-releasing hormone receptor (GnRHR), a unique G-protein-coupled receptor (GPCR) lacking an intracellular carboxyl tail (C-tail), does not follow a ␤-arrestin-dependent internalization pathway

  • ␤-Arrestin-independent Internalization of the GnRHR/cf Tail Chimera—We have previously shown that internalization of the rat GnRHR is not affected by overexpression of ␤-arrestin in COS cells or by the high endogenous levels of ␤-arrestin when expressed in HEK 293 cells [1]

Read more

Summary

The abbreviations used are

GnRHR, gonadotropin-releasing hormone receptor; GPCR, G-protein-coupled receptor; CKII, casein kinase II; TRH, thyrotropin-releasing hormone; TRHR, TRH receptor; GRK, G-protein coupled receptor kinase; PKC, protein kinase C; C-tail, carboxyl-terminal tail; HEK, human embryonic kidney; cf, catfish; cfGnRHR, catfish GnRHR; HA, hemagglutinin; IP, inositol phosphate; GFP, green fluorescent protein; WT, wild type. We observed that chimeric GnRH receptors with extended C-tails containing CKII consensus sites gained ␤-arrestin-dependence, whereas GnRH receptors with C-tails lacking these sites remained ␤-arrestin-insensitive This prompted us to investigate the role of CKII sites in GPCR regulation. This study provides evidence for a novel role of C-terminally located CKII sites in determining the sensitivity of a GPCR to internalize via the ␤-arrestin-dependent pathway

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