Abstract

The G protein-coupled protease-activated receptor 1 (PAR1) is irreversibly proteolytically activated by thrombin. Hence, the precise regulation of PAR1 signaling is important for proper cellular responses. In addition to desensitization, internalization and lysosomal sorting of activated PAR1 are critical for the termination of signaling. Unlike most G protein-coupled receptors, PAR1 internalization is mediated by the clathrin adaptor protein complex 2 (AP-2) and epsin-1, rather than β-arrestins. However, the function of AP-2 and epsin-1 in the regulation of PAR1 signaling is not known. Here, we report that AP-2, and not epsin-1, regulates activated PAR1-stimulated phosphoinositide hydrolysis via two different mechanisms that involve, in part, a subset of R4 subfamily of "regulator of G protein signaling" (RGS) proteins. A significantly greater increase in activated PAR1 signaling was observed in cells depleted of AP-2 using siRNA or in cells expressing a PAR1 (420)AKKAA(424) mutant with defective AP-2 binding. This effect was attributed to AP-2 modulation of PAR1 surface expression and efficiency of G protein coupling. We further found that ectopic expression of R4 subfamily members RGS2, RGS3, RGS4, and RGS5 reduced activated PAR1 wild-type signaling, whereas signaling by the PAR1 AKKAA mutant was minimally affected. Intriguingly, siRNA-mediated depletion analysis revealed a function for RGS5 in the regulation of signaling by the PAR1 wild type but not the AKKAA mutant. Moreover, activation of the PAR1 wild type, and not the AKKAA mutant, induced Gαq association with RGS3 via an AP-2-dependent mechanism. Thus, AP-2 regulates activated PAR1 signaling by altering receptor surface expression and through recruitment of RGS proteins.

Highlights

  • The function of the clathrin adaptor adaptor protein complex 2 (AP-2) in the regulation of G protein-coupled receptors (GPCRs) coupling to G protein signaling is not known

  • Only coexpression of RGS3 was observed to cause a modest but significant decrease in protease-activated receptor 1 (PAR1) AKKAA mutant signaling, its overall effect was reduced substantially compared with its effect at wild-type PAR1 (Fig. 6, B and C). These findings suggest that RGS2, RGS3, RGS4, and RGS5 function as negative regulators of activated PAR1-induced PI signaling, whereas the PAR1 AKKAA mutant with defective AP-2 interaction is less sensitive to regulator of G protein signaling” (RGS) protein regulation of agonist-stimulated G protein signaling

  • GPCR signaling is precisely regulated through various mechanisms mediated by ␤-arrestins, which function to uncouple the receptor from G protein signaling and promote receptor internalization [29]

Read more

Summary

Background

The function of the clathrin adaptor AP-2 in the regulation of GPCR coupling to G protein signaling is not known. Unlike most G protein-coupled receptors, PAR1 internalization is mediated by the clathrin adaptor protein complex 2 (AP-2) and epsin-1, rather than ␤-arrestins. AP-2 regulates activated PAR1 signaling by altering receptor surface expression and through recruitment of RGS proteins. Our findings suggest that AP-2 functions as a critical regulator of PAR1 signaling activity both by modulating receptor surface expression and through recruitment of a subset of the R4 family of RGS proteins. These findings reveal a novel role for AP-2 in the regulation of RGS protein recruitment to G proteins for certain GPCRs

EXPERIMENTAL PROCEDURES
RESULTS
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