Abstract

Mutations to the adhesion G protein-coupled receptor ADGRG1 (G1; also known as GPR56) underlie the neurological disorder bilateral frontoparietal polymicrogyria. Disease-associated mutations in G1 studied to date are believed to induce complete loss of receptor function through disruption of either receptor trafficking or signaling activity. Given that N-terminal truncation of G1 and other adhesion G protein-coupled receptors has been shown to significantly increase the receptors' constitutive signaling, we examined two different bilateral frontoparietal polymicrogyria-inducing extracellular loop mutations (R565W and L640R) in the context of both full-length and N-terminally truncated (ΔNT) G1. Interestingly, we found that these mutations reduced surface expression of full-length G1 but not G1-ΔNT in HEK-293 cells. Moreover, the mutations ablated receptor-mediated activation of serum response factor luciferase, a classic measure of Gα12/13-mediated signaling, but had no effect on G1-mediated signaling to nuclear factor of activated T cells (NFAT) luciferase. Given these differential signaling results, we sought to further elucidate the pathway by which G1 can activate NFAT luciferase. We found no evidence that ΔNT activation of NFAT is dependent on Gαq/11-mediated or β-arrestin-mediated signaling but rather involves liberation of Gβγ subunits and activation of calcium channels. These findings reveal that disease-associated mutations to the extracellular loops of G1 differentially alter receptor trafficking, depending on the presence of the N terminus, and differentially alter signaling to distinct downstream pathways.

Highlights

  • Adhesion G protein-coupled receptors2 are a family of GPCRs comprising 33 receptors in humans

  • The ⌬NT versions of G protein-coupled receptor ADGRG1 (G1) lack most of the N terminus (⌬ amino acids 1–382) up to the site of predicted GPCR autoproteolysis-inducing (GAIN) domain cleavage and mimic the C-terminal fragment (CTF) of G1 that is cleaved at the GAIN domain and undergoes dissociation from the Nterminal fragment (NTF)

  • Given that the R565W and L640R mutations disrupted signaling to serum response factor (SRF) luciferase but preserved signaling to nuclear factor of activated T cells (NFAT) luciferase and interaction with ␤-arrestins, we explored whether ␤-arrestins might be involved in mediating G1 signaling to NFAT luciferase

Read more

Summary

To whom correspondence should be addressed

Numerous aspects of normal physiology as well as pathological processes [1]. Adhesion GPCRs possess extremely long extracellular N termini that are often decorated with multiple protein/protein interaction domains. Studies by several groups have delineated a model of activation termed the cryptic agonist model wherein dissociation of the NTF from the membrane-embedded CTF unveils the agonistic properties of the remaining extracellular stalk ( termed the “stachel”) (9 –11, 34). We previously investigated this model for G1 and found that the stalk is essential for some but not all signaling outputs [35]. For other aGPCRs, such as ADGRB1 (BAI1), the presence of the extracellular stalk does not appear to matter at all for receptor signaling activity [35]. The results of these studies have provided new insights into the regulation of G1 signaling and the mechanisms by which these mutations cause human disease

Results
Discussion
Experimental procedures
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