Associate editor: V. Watts
Physiological and pharmacological implications of beta-arrestin regulation

https://doi.org/10.1016/j.pharmthera.2008.11.005Get rights and content

Abstract

G protein-coupled receptor-targeted drug discovery as well as “compound reassessment” requires the utilization of diverse screens to determine agonist efficacies and potencies beyond the scope of ligand binding and G protein coupling. Such efforts have arisen from extensive studies, both in cellular and animal models, demonstrating that these seven transmembrane domain-spanning, G protein-coupled receptors may engage in more diverse functions than their name suggests and particular focus is drawn to their interactions with beta-arrestins (βarrestins). As regulators, βarrestins are involved in dampening G protein-coupling pathways. βArrestins can also play pro-signaling roles in receptor mediated events and the coupling of receptors to βarrestins may be as important as their potential to couple to G proteins in the physiological setting. In the last decade, the development of βarrestin deficient mouse models has allowed for the assessment of the contribution of individual βarrestins to receptor function in vivo. This review will discuss the current literature that implicates βarrestins in receptor function in respect to physiological and behavioral responses observed in the live animal model.

Introduction

βArrestins (non-visual arrestins) are ubiquitously expressed proteins that were first described for their role in desensitizing G protein-coupled receptors (GPCRs). There are two βarrestins, namely, βarrestin1 and βarrestin2, which are also referred to as arrestin-2 and arrestin-3, respectively. As their names imply, βarrestins were first identified for their ability to “arrest” agonist-stimulated β2 adrenergic receptor (β2AR) signaling (Lohse et al., 1990) in a manner similar to arrestin regulation of rhodopsin. The canonical model of GPCR regulation by βarrestins also involves GPCR kinases (GRKs) which phosphorylate receptors and thereby serve to facilitate receptor-βarrestin interactions (Benovic et al., 1987, Sibley et al., 1987, Lohse et al., 1992, Pitcher et al., 1992). Upon complexing with receptors, βarrestins can serve as inhibitors of signal transduction by preventing further receptor coupling to G protein signaling cascades (for reviews see: Premont et al., 1995, Freedman and Lefkowitz, 1996, Lefkowitz, 1998).

Specific examples of βarrestins serving as negative regulators of GPCR signaling are plentiful in cellular as well as animal model systems (Table 1) (for reviews see: Gainetdinov et al., 2004, Bohn et al., 2004a, Gurevich and Gurevich, 2006, Premont and Gainetdinov, 2007). In addition to mediating receptor desensitization, βarrestins can facilitate recruitment and interactions between GPCRs and signaling partners. In this capacity, βarrestins can serve as positive mediators of receptor signaling to downstream targets. Evidence for GPCRs coupling to βarrestins to transduce receptor signaling has also been widely demonstrated in cellular models (for reviews see: Luttrell et al., 1999, Luttrell, 2002, Lefkowitz and Shenoy, 2005, DeWire et al., 2007). Studies in mouse models also support a pro-signaling role for βarrestins (particularly βarrestin2) and these reports are summarized in Table 2.

Arguably, the most studied GPCR is the β2AR. In vitro studies with this receptor have been instrumental in demonstrating the diverse and pleiotropic roles that βarrestins can play in determining agonist-induced receptor responses. The β2AR has been shown to interact with both βarrestin1 and βarrestin2 upon agonist stimulation (Attramadal et al., 1992) and such interactions result in decreased responsiveness to agonist over time. The removal of βarrestins by early anti-sense studies (Mundell et al., 1999), later siRNA studies (Ahn et al., 2003), as well as studies utilizing mouse embryonic fibroblasts devoid of both βarrestin1 and βarrestin2 (Kohout et al., 2001), confirm that βarrestins play a critical role in promoting this waning effect on G protein-coupling and adenylyl cyclase stimulation following agonist activation of the β2AR. Similar studies have been performed for multiple GPCRs of diverse classes and together, these findings support the canonical model wherein the agonist-activated GPCR becomes phosphorylated by GRKs which subsequently increases the binding affinity of the receptor for βarrestins.

βArrestin interactions with activated GPCRs can be detected by co-immunoprecipitation (Groer et al., 2007), confocal microscopy (Barak et al., 1997), bioluminescence resonance energy transfer (BRET) (Hamdan et al., 2005), and fluorescence resonance energy transfer (FRET) (Drake et al., 2008) assays. Such developments, including enzyme complementation assays (von Degenfeld et al., 2007), have facilitated high throughput screens for assessing drug-induced βarrestin-receptor interactions. Looking forward, the interactions between βarrestins and GPCRs may be realized for ultimately determining relative drug efficacies in vivo (Claing and Laporte, 2005, Violin and Lefkowitz, 2007, DeWire et al., 2007).

Section snippets

βArrestin regulation of GPCRs in vivo

While cellular model systems have been particularly useful for determining which receptors can possibly interact with βarrestins, in many cases, the question remains as to whether such interactions are pharmacologically and physiologically relevant. Assessing βarrestin function in vivo is challenging as there are no selective inhibitors of βarrestins. Some attempts have been made to develop selective inhibitors to GRKs as a means to prevent subsequent βarrestin recruitment, yet the degree of

β2 Adrenergic receptor regulation

Given the considerable evidence demonstrating βarrestin-mediated regulation of the β2AR, it is not surprising that the initial studies were focused on unveiling a cardiac phenotype in these animals. While the βarr1-KO mice present no overt gross phenotypes, they do display an altered response to β2AR agonist challenge (Conner et al., 1997). In this study, heart rates and ejection fractions between anesthetized wildtype (WT) and βarr1-KO mice were not different, as assessed by echocardiography;

βArrestin2 knockout mice

The original colony of βarrestin2 knockout (βarr2-KO) mice was generated in a C57Bl/6 X 129 SvJ mixed mouse strain (Bohn et al., 1999) and has subsequently been backcrossed onto a congenic C57Bl/6 strain. Both the original mixed strain and the backcrossed strain are used in current studies. There are no overt gross phenotypes that distinguish βarr2-KO mice from their WT counterparts. However, there are subtle differences between the two genotypes that have been found upon closer evaluation of

Summary

Cell model characterizations have been critical for determining which proteins can interact to influence cellular function, and studies of this nature have opened new avenues for considering drug function in respect to receptor-pathway engagement. Although receptor-βarrestin interactions may be observed in cells, the role that βarrestins play in vivo, wherein receptor levels may be quite low and GRK levels may vary between tissues, may be difficult to predict. The use of genetically modified

References (77)

  • KenakinT.

    Collateral efficacy in drug discovery: taking advantage of the good (allosteric) nature of 7TM receptors

    Trends Pharmacol Sci

    (2007)
  • KiefferB.L.

    Opioids: first lessons from knockout mice

    Trends Pharmacol Sci

    (1999)
  • KiefferB.L. et al.

    Exploring the opioid system by gene knockout

    Prog Neurobiol

    (2002)
  • LeBarsD. et al.

    Effects of morphine upon the lamina V type cells activities in the dorsal horn of the decerebrate cat

    Brain Res

    (1976)
  • LefkowitzR.J.

    G protein-coupled receptors. III. New roles for receptor kinases and beta-arrestins in receptor signaling and desensitization

    J Biol Chem

    (1998)
  • LohseM.J. et al.

    Receptor-specific desensitization with purified proteins. Kinase dependence and receptor specificity of beta-arrestin and arrestin in the beta 2-adrenergic receptor and rhodopsin systems

    J Biol Chem

    (1992)
  • LuttrellL.M. et al.

    Regulation of tyrosine kinase cascades by G-protein-coupled receptors

    Curr Opin Cell Biol

    (1999)
  • MailmanR.B.

    GPCR functional selectivity has therapeutic impact

    Trends Pharmacol Sci

    (2007)
  • PrzewlockaB. et al.

    Knockdown of spinal opioid receptors by antisense targeting beta-arrestin reduces morphine tolerance and allodynia in rat

    Neurosci Lett

    (2002)
  • SchmidtC.J. et al.

    The 5-HT2 receptor antagonist, MDL 28,133A, disrupts the serotonergic-dopaminergic interaction mediating the neurochemical effects of 3,4-methylenedioxymethamphetamine

    Eur J Pharmacol

    (1992)
  • ShenoyS.K. et al.

    beta-arrestin-dependent, G protein-independent ERK1/2 activation by the beta2 adrenergic receptor

    J Biol Chem

    (2006)
  • SibleyD.R. et al.

    Regulation of transmembrane signaling by receptor phosphorylation

    Cell

    (1987)
  • TohgoA. et al.

    beta-Arrestin scaffolding of the ERK cascade enhances cytosolic ERK activity but inhibits ERK-mediated transcription following angiotensin AT1a receptor stimulation

    J Biol Chem

    (2002)
  • ViolinJ.D. et al.

    Beta-arrestin-biased ligands at seven-transmembrane receptors

    Trends Pharmacol Sci

    (2007)
  • AhnS. et al.

    Desensitization, internalization, and signaling functions of beta-arrestins demonstrated by RNA interference

    Proc Natl Acad Sci USA

    (2003)
  • ArlindeC. et al.

    A cluster of differentially expressed signal transduction genes identified by microarray analysis in a rat genetic model of alcoholism

    Pharmacogenomics J

    (2004)
  • BenovicJ.L. et al.

    Functional desensitization of the isolated beta-adrenergic receptor by the beta-adrenergic receptor kinase: potential role of an analog of the retinal protein arrestin (48-kDa protein)

    Proc Natl Acad Sci USA

    (1987)
  • BjorkK. et al.

    Modulation of voluntary ethanol consumption by beta-arrestin 2

    Faseb J

    (2008)
  • BohnL.M. et al.

    Relative opioid efficacy is determined by the complements of the G protein-coupled receptor desensitization machinery

    Mol Pharmacol

    (2004)
  • BohnL.M. et al.

    G protein-coupled receptor kinase/beta-arrestin systems and drugs of abuse: psychostimulant and opiate studies in knockout mice

    Neuromolecul Med

    (2004)
  • BohnL.M. et al.

    Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence

    Nature

    (2000)
  • BohnL.M. et al.

    Enhanced rewarding properties of morphine, but not cocaine, in beta(arrestin)-2 knock-out mice

    J Neurosci

    (2003)
  • BohnL.M. et al.

    Differential mechanisms of morphine antinociceptive tolerance revealed in (beta)arrestin-2 knock-out mice

    J Neurosci

    (2002)
  • BohnL.M. et al.

    Enhanced morphine analgesia in mice lacking beta-arrestin 2

    Science

    (1999)
  • BreivogelC.S. et al.

    Sensitivity to delta9-tetrahydrocannabinol is selectively enhanced in beta-arrestin2−/− mice

    Behav Pharmacol

    (2008)
  • ChausmerA.L. et al.

    The role of D2-like dopamine receptors in the locomotor stimulant effects of cocaine in mice

    Psychopharmacology (Berl)

    (2001)
  • ClaingA. et al.

    Novel roles for arrestins in G protein-coupled receptor biology and drug discovery

    Curr Opin Drug Discov Dev

    (2005)
  • ConnerD.A. et al.

    beta-Arrestin1 knockout mice appear normal but demonstrate altered cardiac responses to beta-adrenergic stimulation

    Circ Res

    (1997)
  • Cited by (89)

    View all citing articles on Scopus
    View full text