Design and functional characterization of a novel, arrestin-biased designer G protein-coupled receptor

K Nakajima, J Wess - Molecular pharmacology, 2012 - ASPET
Molecular pharmacology, 2012ASPET
Mutational modification of distinct muscarinic receptor subtypes has yielded novel designer
G protein-coupled receptors (GPCRs) that are unable to bind acetylcholine (ACh), the
endogenous muscarinic receptor ligand, but can be efficiently activated by clozapine-N-
oxide (CNO), an otherwise pharmacologically inert compound. These CNO-sensitive
designer GPCRs [alternative name: designer receptors exclusively activated by designer
drug (DREADDs)] have emerged as powerful new tools to dissect the in vivo roles of distinct …
Mutational modification of distinct muscarinic receptor subtypes has yielded novel designer G protein-coupled receptors (GPCRs) that are unable to bind acetylcholine (ACh), the endogenous muscarinic receptor ligand, but can be efficiently activated by clozapine-N-oxide (CNO), an otherwise pharmacologically inert compound. These CNO-sensitive designer GPCRs [alternative name: designer receptors exclusively activated by designer drug (DREADDs)] have emerged as powerful new tools to dissect the in vivo roles of distinct G protein signaling pathways in specific cell types or tissues. As is the case with other GPCRs, CNO-activated DREADDs not only couple to heterotrimeric G proteins but can also recruit proteins of the arrestin family (arrestin-2 and -3). Accumulating evidence suggests that arrestins can act as scaffolding proteins to promote signaling through G protein-independent signaling pathways. To explore the physiological relevance of these arrestin-dependent signaling pathways, the availability of an arrestin-biased DREADD would be highly desirable. In this study, we describe the development of an M3 muscarinic receptor-based DREADD [Rq(R165L)] that is no longer able to couple to G proteins but can recruit arrestins and promote extracellular signal-regulated kinase-1/2 phosphorylation in an arrestin- and CNO-dependent fashion. Moreover, CNO treatment of mouse insulinoma (MIN6) cells expressing the Rq(R165L) construct resulted in a robust, arrestin-dependent stimulation of insulin release, directly implicating arrestin signaling in the regulation of insulin secretion. This newly developed arrestin-biased DREADD represents an excellent novel tool to explore the physiological relevance of arrestin signaling pathways in distinct tissues and cell types.
ASPET