Elsevier

Cellular Signalling

Volume 80, April 2021, 109905
Cellular Signalling

Biased agonism at β-adrenergic receptors

https://doi.org/10.1016/j.cellsig.2020.109905Get rights and content

Highlights

  • G protein and β-arrestin biased agonists have been characterized for the βARs.

  • Biased β-agonists may improve clinical outcomes compared to balanced β-agonists.

  • TM VII / ECL movements and GRK interaction are implicated in bias mechanism.

  • Biased drug discovery efforts and experimental design require careful consideration.

Abstract

The β-adrenergic receptors (βARs) include three subtypes, β1, β2 and β3. These receptors are widely expressed and regulate numerous physiological processes including cardiovascular and metabolic functions and airway tone. The βARs are also important targets in the treatment of many diseases including hypertension, heart failure and asthma. In some cases, the use of current βAR ligands to treat a disease is suboptimal and can lead to severe side effects. One strategy to potentially improve such treatments is the development of biased agonists that selectively regulate a subset of βAR signaling pathways and responses. Here we discuss the compounds identified to date that preferentially activate a Gs- or β-arrestin-mediated signaling pathway through βARs. Mechanistic insight on how these compounds bias signaling sheds light on the potential development of even more selective compounds that should have increased utility in treating disease.

Introduction

The β-adrenergic receptors (βARs) are a subfamily of G protein-coupled receptors (GPCRs) that are expressed by most cell types in humans [1]. This subfamily consists of three members, β1, β2, and β3AR, and are the targets of the endogenous catecholamines epinephrine and norepinephrine [2,3]. Signaling through βARs regulates a wide variety of physiological processes including cardiac function, airway tone, metabolic function, and others [4]. Due to their ubiquity and key role in human health, the βARs are cornerstone drug targets for a variety of pathologies [5], and drug discovery efforts around these receptors have generated a diverse set of pharmacological agents [6].

Canonically, β-agonists promote receptor mediated G protein activation primarily through Gs to activate the enzyme adenylyl cyclase and increase cAMP production. While there are subtype differences, activated βARs are typically phosphorylated by regulatory kinases such as GPCR kinases (GRKs), and signaling is then terminated via interaction with β-arrestins, a process called desensitization [7]. In recent years, it has become widely accepted that this cycle is an incomplete description of the signaling repertoire of the βARs. An additional role of β-arrestins as signal transducers has been identified [8] and β-arrestins can also mediate receptor internalization (Fig. 1A). While regulation of β3AR signaling is less well characterized than β1AR or β2AR, the β3AR does not interact with GRKs or β-arrestins and desensitization is observed more often after hours to days rather than minutes [9]. This is due in part to the lack of regulatory kinase phosphorylation sites on the C-terminal tail of the β3AR that are found in the β1AR and β2AR. Evidence suggests that regulation of β3AR involves downregulation of its mRNA and the receptor protein itself, although this varies across the systems tested and appears to be cell type dependent [9] (Fig. 1B). In addition, βARs have been shown to couple to multiple G proteins, and G protein and β-arrestin interaction with the receptor can be selectively promoted by ligands that stabilize distinct receptor conformations. The selective activation of these pathways is known as biased signaling (Fig. 2). In the early 2000s, the concepts of “pluridimensional efficacy” and “ligand-biased signaling” were first observed for the β2AR when compounds that were previously characterized as receptor antagonists were reported to have the ability to stimulate β-arrestin-dependent MAP kinase signaling [10] while subsequent studies pioneered the re-classification of β2AR ligands [11,12]. This review will focus on recent developments in this signaling paradigm at the βARs.

Section snippets

The βAR subfamily

The βAR subfamily includes the β1, β2, and β3ARs. These receptors are generally highly conserved within the transmembrane domains that mediate ligand binding and ligand-induced conformational changes while the extracellular and intracellular regions are poorly conserved (Fig. 3). These receptors coordinate physiological responses to the catecholamines epinephrine and norepinephrine, which promote activation of their cognate G protein, Gs. In recent years, an expanded view of the βAR interactome

Clinical utility of biased signaling at βARs

Arrestin biased signaling at the β1AR provides additional clinical utility compared to balanced antagonists for cardiopathies. Compensatory dysregulation of the sympathetic nervous system causes an increase in circulating catecholamines in compensated, stable congestive heart failure. Blocking G protein signaling through cardiac β1AR has a reductive effect on the heart rate and ultimately leads to an improved ejection fraction. A β-arrestin biased agonist improves on this therapeutic strategy

Arrestin biased β-agonists

The compounds described below represent a selection of reported β-arrestin biased β-agonists. This series of compounds is not necessarily exhaustive, but is a cross-section of compounds demonstrating this signaling phenotype. The structures of the small molecules are presented in Fig. 4A, and pharmacological activity is summarized in Table 1.

G protein biased β-agonists

The compounds described below represent a non-exhaustive cross-section of compounds that demonstrate a G protein biased signaling phenotype through βARs. The structures of the small molecules are presented in Fig. 4B, and pharmacological activity is summarized in Table 1.

Mechanistic insights into βAR biased signaling

Studies with several different GPCRs suggest that transmembrane (TM) VII mediates signaling bias and receptor coupling to β-arrestin [[65], [66], [67], [68]]. A very recent study examined the role of TM VII of the β2AR using an in vitro single molecule fluorescence system to examine the role of conformational exchange kinetics on β-arrestin bias [69]. In this study, a Cy3 fluorophore was chemically conjugated to a cysteine residue on TM VII. Using the agonist formoterol and comparatively

Limitations

Despite the clear therapeutic potential of biased agonism at the βARs, this class of compounds has had limited clinical success [35,75]. The difficulty in translating biased pharmacological agents from the laboratory to medical practice highlights the challenges intrinsic to this therapeutic strategy. These obstacles and strategies to mitigate them are thoroughly explored in recent reviews [76,77]. To summarize briefly, there are several explanations for why molecules that demonstrate the

Conclusions

Biased signaling at βARs exemplifies the complexities of GPCR pharmacology. Our increased understanding of the molecular mechanisms of βAR signaling will allow for the development of better therapeutics with fewer side effects. Leveraging these developments will be an important aspect in facilitating the next generation of βAR ligands to generate more precise physiological responses. The popularity of the βARs as model systems for GPCR signaling paradigms has led to increases in the

Declaration of Competing Interest

The authors declare no competing interests.

Acknowledgements

This work was supported in part by National Institutes of Health grants R35 GM122541, R01 HL136219 and P01 HL114471 (to JLB) and T32 GM100836 and F31 HL139104 (to MI). Fig. 1, Fig. 2 were created with BioRender.com.

References (80)

  • S.K. Shenoy

    β-arrestin-dependent, G protein-independent ERK1/2 activation by the β2 adrenergic receptor

    J. Biol. Chem.

    (2006)
  • T. Münzel et al.

    Nebivolol: the somewhat-different beta-adrenergic receptor blocker

    J. Am. Coll. Cardiol.

    (2009)
  • R. Carr

    Development and characterization of pepducins as Gs-biased allosteric agonists

    J. Biol. Chem.

    (2014)
  • H. Ikram et al.

    Xamoterol in severe heart failure

    Lancet

    (1990)
  • M. Bermudez et al.

    Does divergent binding pocket closure drive ligand bias for class A GPCRs?

    Trends Pharmacol. Sci.

    (2019)
  • R.T. Strachan

    Divergent transducer-specific molecular efficacies generate biased agonism at a G protein-coupled receptor (GPCR)

    J. Biol. Chem.

    (2014)
  • M. Bermudez et al.

    Strategies for the discovery of biased GPCR ligands

    Drug Discov. Today

    (2019)
  • A.D. Strosberg

    Structure, function, and regulation of adrenergic receptors

    Protein Sci.

    (1993)
  • L.J. Emorine

    The human β3-adrenergic receptor: relationship with atypical receptors

    Am. J. Clin. Nutr.

    (1992)
  • P.A. Insel

    Adrenergic receptors - evolving concepts and clinical implications

    N. Engl. J. Med.

    (1996)
  • C.B. Patel et al.

    Functional selectivity in adrenergic and angiotensin signaling systems

    Mol. Pharmacol.

    (2010)
  • S.Y. Hodavance et al.

    G protein-coupled receptor biased agonism

    J. Cardiovasc. Pharmacol.

    (2016)
  • R.J. Lefkowitz et al.

    Transduction of receptor signals by β-arrestins

    Science

    (2007)
  • K. Okeke et al.

    Agonist-induced desensitisation of β3-adrenoceptors: where, when, and how?

    Br. J. Pharmacol.

    (2019)
  • M. Azzi

    β-arrestin-mediated activation of MAPK by inverse agonists reveals distinct active conformations for G protein-coupled receptors

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • E.T. van der Westhuizen et al.

    Quantification of ligand bias for clinically relevant β2-adrenergic receptor ligands: implications for drug taxonomy

    Mol. Pharmacol.

    (2014)
  • J.R. Helper et al.

    G proteins

    Trends Biochem. Sci.

    (1992)
  • K.N. Nobles

    Distinct phosphorylation sites on the β2-adrenergic receptor establish a barcode that encodes differential functions of β-arrestin

    Sci. Signal.

    (2011)
  • A. Musa Zamah et al.

    Protein kinase A-mediated phosphorylation of the β2-adrenergic receptor regulates its coupling to Gs and Gi: demonstration in a reconstituted system

    J. Biol. Chem.

    (2002)
  • Y. Daaka et al.

    Switching of the coupling of the β2-adrenergic receptor to different G proteins by protein kinase A

    Nature

    (1997)
  • S.S.G. Ferguson

    Role of β-arrestin in mediating agonist-promoted G protein-coupled receptor internalization

    Science

    (1996)
  • O.B. Goodman

    β-arrestin acts as a clathrin adaptor in endocytosis of the β2-adrenergic receptor

    Nature

    (1996)
  • S.M. DeWire et al.

    Beta-arrestins and cell signaling

    Annu. Rev. Physiol.

    (2007)
  • M. O'Hayre

    Genetic evidence that β-arrestins are dispensable for the initiation of β2-adrenergic receptor signaling to ERK

    Sci. Signal.

    (2017)
  • M. Grundmann

    Lack of beta-arrestin signaling in the absence of active G proteins

    Nat. Commun.

    (2018)
  • L.M. Luttrell

    Manifold roles of β-arrestins in GPCR signaling elucidated with siRNA and CRISPR/Cas9

    Sci. Signal.

    (2018)
  • G. Schena et al.

    Everything you always wanted to know about β3-AR* (*but were afraid to ask)

    Cells

    (2019)
  • I.M. Kim

    β-blockers alprenolol and carvedilol stimulate β-arrestin- mediated EGFR transactivation

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • Q. Wang

    Carvedilol induces biased β1 adrenergic receptor-nitric oxide synthase 3-cyclic guanylyl monophosphate signaling to promote cardiac contractility

    Cardiovasc. Res.

    (2020)
  • X. Liu

    β-Arrestin-biased signaling mediates memory reconsolidation

    Proc. Natl. Acad. Sci. U. S. A.

    (2015)
  • Cited by (0)

    View full text