Spinophilin as a novel regulator of M3R -mediated insulin release Recently, we tested the hypothesis that spinophilin (SPL), a multi-domain scaffolding protein, might play a role in modulating the activity of eta-cell M3Rs. This study was prompted by the observation that SPL can regulate the activity of various CNS functions mediated by distinct GPCRs. We demonstrated, by using both in vitro and in vivo approaches (mouse insulinoma cells and SPL-deficient mice), that SPL is a potent negative regulator of M3R-mediated signaling and insulin release. Additional biochemical and biophysical studies, including the use of BRET technology, suggested that SPL is able to recruit RGS4 to the M3R signaling complex in an agonist-dependent fashion. Since RGS4 is a member of the RGS family of proteins which act to reduce the lifetime of activated G proteins, these findings support the concept that the inhibitory effects of SPL on M3R activity are mediated by RGS4. This is the first report demonstrating a role for SPL in regulating eta-cell function. Given the importance of beta-cell M3Rs in maintaining normal blood glucose levels, our findings highlight the significant functional role of M3R (GPCR)-associated proteins in modulating eta-cell biology. Such proteins may prove useful as targets for novel classes of drugs aimed at improving eta-cell function for therapeutic purposes. Development of a designer GPCR useful for studying the roles of arrestins in regulating insulin release and other physiological functions Activated GPCRs are rapidly phosphorylated by GPCR kinases (GRKs), promoting interactions between the phosphorylated receptors and members of the arrestin protein family (arrestin-2 and -3), a process which interferes with receptor/G protein coupling. However, during the past decade, it has become increasingly clear that arrestin-2 and -3 can serve as adaptor proteins that transduce signals to multiple effector pathways. Although some progress has been made in delineating the physiological functions of arrestin signaling pathways in various tissues, much remains to be learned about the in vivo physiological relevance of arrestin-mediated signaling. Recently, various mutant muscarinic receptors have been developed 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. To explore the physiological relevance of arrestin-mediated signaling cascades, we recently developed a novel, arrestin-biased CNO-sensitive designer receptor (Rq(R165L)) as a novel experimental tool. This M3 receptor-based DREADD was no longer able to couple to G proteins but could recruit arrestins and promote ERK1/2 phosphorylation in an arrestin- and CNO-dependent fashion. Moreover, CNO treatment of MIN6 insulinoma 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.

Project Start
Project End
Budget Start
Budget End
Support Year
4
Fiscal Year
2012
Total Cost
$1,824,750
Indirect Cost
City
State
Country
Zip Code
Zhu, Lu; Rossi, Mario; Cui, Yinghong et al. (2017) Hepatic ?-arrestin 2 is essential for maintaining euglycemia. J Clin Invest 127:2941-2945
Zhu, Lu; Almaça, Joana; Dadi, Prasanna K et al. (2017) ?-arrestin-2 is an essential regulator of pancreatic ?-cell function under physiological and pathophysiological conditions. Nat Commun 8:14295
Nakajima, Ken-ichiro; Cui, Zhenzhong; Li, Chia et al. (2016) Gs-coupled GPCR signalling in AgRP neurons triggers sustained increase in food intake. Nat Commun 7:10268
Wess, Jürgen (2016) Use of Designer G Protein-Coupled Receptors to Dissect Metabolic Pathways. Trends Endocrinol Metab 27:600-603
Klepac, Katarina; Kili?, Ana; Gnad, Thorsten et al. (2016) The Gq signalling pathway inhibits brown and beige adipose tissue. Nat Commun 7:10895
Rossi, Mario; Cui, Zhenzhong; Nakajima, Ken-Ichiro et al. (2015) Virus-Mediated Expression of DREADDs for In Vivo Metabolic Studies. Methods Mol Biol 1335:205-21
Rossi, Mario; Ruiz de Azua, Inigo; Barella, Luiz F et al. (2015) CK2 acts as a potent negative regulator of receptor-mediated insulin release in vitro and in vivo. Proc Natl Acad Sci U S A 112:E6818-24
Nakajima, Kenichiro; Jain, Shalini; Ruiz de Azua, Inigo et al. (2013) Minireview: Novel aspects of M3 muscarinic receptor signaling in pancreatic ýý-cells. Mol Endocrinol 27:1208-16
Wess, Jürgen; Nakajima, Kenichiro; Jain, Shalini (2013) Novel designer receptors to probe GPCR signaling and physiology. Trends Pharmacol Sci 34:385-92
Jain, Shalini; Ruiz de Azua, Inigo; Lu, Huiyan et al. (2013) Chronic activation of a designer G(q)-coupled receptor improves ? cell function. J Clin Invest 123:1750-62

Showing the most recent 10 out of 24 publications