Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Curr Opin Endocr Metab Res. Author manuscript; available in PMC 2022 Feb 1.
Published in final edited form as:
PMCID: PMC7945687
NIHMSID: NIHMS1630195
PMID: 33718657

Targeting G protein-coupled receptor kinases (GRKs) to G protein-coupled receptors

Abstract

G protein-coupled receptors (GPCRs) interact with three protein families following agonist binding: heterotrimeric G proteins, G protein-coupled receptor kinases (GRKs) and arrestins. GRK-mediated phosphorylation of GPCRs promotes arrestin binding to uncouple the receptor from G protein, a process called desensitization, and for many GPCRs, arrestin binding also promotes receptor endocytosis and intracellular signaling. Thus, GRKs play a central role in modulating GPCR signaling and localization. Here we review recent advances in this field which include additional insight into how GRKs target GPCRs and bias signaling, and the development of specific inhibitors to dissect GRK function in model systems.

Keywords: arrestin, cell signaling, desensitization, endocytosis, phosphorylation, protein kinase

Introduction

G protein-coupled receptors (GPCRs) transmit extracellular stimuli into intracellular responses via their ability to activate heterotrimeric G proteins. GPCR signaling is then modulated by agonist-dependent phosphorylation of the receptor by G protein-coupled receptor kinases (GRKs) [1]. This promotes arrestin binding which inhibits G protein coupling, a process termed desensitization. Non-visual arrestins (β-arrestins) also initiate receptor endocytosis where the active GPCR can continue to signal via G protein- and/or arrestin-dependent pathways. The role of GRKs in regulating GPCR signaling has been extensively studied while aberrant GRK activity has been attributed to a range of disease pathologies and serves as a viable target for intervention [2]. This review provides a concise overview of GRKs with an update on notable advances in GRK function and regulation of GPCRs over the past few years (Figure 1).

An external file that holds a picture, illustration, etc.
Object name is nihms-1630195-f0001.jpg

Overview of GRK targeting to GPCRs and nodes of GRK regulation. (1) GRKs are recruited to agonist-activated GPCRs at the plasma membrane. (2) Upon binding GPCRs, GRKs undergo a conformational change that promotes kinase activation. (3) The active GRK phosphorylates the GPCR C-terminus. (4) GPCR phosphorylation by GRKs promotes β-arrestin (βarr) recruitment to the receptor, mediating receptor desensitization, endocytosis and arrestin-dependent signaling pathways. GRKs and GRK regulation can play a key role in determining receptor signaling. Recruitment factors such as G protein subunits or acidic lipids can mediate GRK recruitment to the receptor. Regulatory factors can directly bind to or post-translationally modify GRKs to regulate kinase recruitment and activity. Crosstalk receptors or the activated GPCR can signal these recruitment or regulatory factors to modulate GRK function. GRK inhibitors block kinase activity, inhibiting GPCR phosphorylation and phosphorylation-dependent pathways.

GRK Family

GRKs were initially discovered in the early 1970s when rhodopsin kinase (now GRK1) was first identified as the kinase responsible for phosphorylating photoactivated rhodopsin [3]. Next, the β-adrenergic receptor kinase (βARK, now GRK2) was identified as a kinase that phosphorylates the active β-adrenergic receptor (βAR) and suggested to be a part of a larger family of GPCR-specific kinases [4]. Following the cloning of GRK1 and GRK2 and the discovery of βARK2 (now GRK3), sequence analysis revealed that βARKs and GRK1 belong to distinct subfamilies [57]. GRK4 (originally called IT11), GRK5 and GRK6 were next discovered, establishing a third GRK subfamily [810]. Finally, GRK7 was identified as a second member of the GRK1 subfamily although primarily restricted to cone photoreceptor cells [11].

GRKs belong to the AGC kinase superfamily and are characterized by a distinct regulator of G protein signaling homology (RH) domain that flanks the kinase domain and regulates GPCR phosphorylation (Figure 2). The RH domain is composed of a terminal subdomain, whose peptide sequence feeds into the kinase N-lobe, and a bundle subdomain that forms intramolecular contacts with the kinase C-lobe. In addition, GRKs contain an N-terminal α-helix (αN-helix) that is critical for kinase function. The GRK C-terminus diverges between subfamilies and mediates lipid binding for membrane localization and GPCR targeting [12]. The GRK1 subfamily is prenylated and the GRK2 subfamily contains a pleckstrin homology (PH) domain that facilitates recruitment to the membrane via interaction with Gβɣ subunits and acidic lipids. Within the GRK4 subfamily, GRK4 and GRK6 are palmitoylated. In addition, a C-terminal amphiphilic α-helix directly mediates GRK5 binding to the plasma membrane and is conserved throughout the GRK4 subfamily [2]. This motif, in conjunction with an N-terminal PIP2 binding site, is predicted to form a basic interface for membrane association [13].

An external file that holds a picture, illustration, etc.
Object name is nihms-1630195-f0002.jpg

GRK domain architecture. GRKs contain a conserved catalytic kinase domain, regulatory N-terminus and regulator of G protein signaling homology (RH) domain. Each GRK subfamily is characterized by a distinct, divergent C-terminal motif that mediates membrane binding. The GRK1 subfamily is prenylated and the GRK2 subfamily contains a PH domain that binds acidic lipids and Gβɣ. At the C-terminus, the GRK4 subfamily is palmitoylated (except for GRK5) and contains a lipid-binding motif. In addition, the GRK4 subfamily has a PIP2 binding site near the N-terminus. Thick cylinders represent defined structural domains. Domains were defined using previously determined structures for GRK5 (PDB ID: 4TNB), GRK6 (PDB ID: 3NYO), GRK2 (PDB ID: 6U7C) and GRK1 (PDB ID: 3C4Z) and GRK1–7 multiple sequence alignment.

GPCR Targeting by GRKs

Previous studies have relied on functional assays to probe GRK regions that mediate GPCR binding, though multiple factors contribute to GRK function at the receptor. Thus, it is difficult to discern whether identified regions are true GPCR binding determinants using these functional readouts. In addition, the relatively low affinity of the GRK/GPCR complex has made traditional techniques such as X-ray crystallography and in vitro binding assays challenging [12]. Two recent studies utilized several innovative methods in parallel to identify key regions responsible for GRK binding to GPCRs (Figure 3A) [13,14]. In the first study, the N- and C-terminal lipid binding sites were identified as regions involved in initial GRK5 recruitment to the β2AR while the GRK5 RH bundle subdomain was also identified as a site of receptor interaction [13]. Interestingly, the N-terminal site is analogous to a region in GRK1 that mediates recruitment to rhodopsin cluster of residues within the GRK RH terminal subdomain was also suggested in GRK1 binding to rhodopsin [14]. This evolutionarily conserved region was previously implicated in regulating GPCR phosphorylation by GRKs and is adjacent to the kinase N-lobe [15]. Therefore, GPCR binding and GRK activity may be intimately related via an allosteric activation network.

An external file that holds a picture, illustration, etc.
Object name is nihms-1630195-f0003.jpg

Structural model of the GRK/GPCR complex and GPCR-mediated GRK activation. Models were adapted from docked β2AR/GRK5 structures based on cross-linking generated by Komolov et al. [13]. (a) Inactive GRK conformation upon recruitment to the active GPCR. The GRK ionic lock establishes contact between the RH and kinase domain to maintain the closed, inactive state. The N-terminal lipid-binding domain (royal blue) [13] and the RH terminal domain (lilac) [14], regions thought to mediate GPCR targeting, are contiguous with the receptor. (b) Domain movements for kinase activation. Arrows indicate GRK domain changes when transitioning from an inactive conformation (as shown) to the active state. Model from (a) is rotated 55° counterclockwise to better visualize domain movements upon activation. The RH domain opens outward and backward from the kinase domain and the kinase N-lobe undergoes closure towards the kinase C-lobe. The entire kinase rotates clockwise relative to the GPCR. (c) GRK active conformation for GPCR phosphorylation.

GPCR/GRK complex formation is coupled to interaction of the GRK kinase domain with the receptor C-terminus for phosphorylation [13]. A recent study introduced a reciprocal relationship where the μ-opioid receptor (MOR) C-terminal phosphorylation sites mediate GRK2 recruitment to the receptor. In addition, a cluster of phosphorylation sites (TSST, residues 354–357) dictate robust downstream multisite phosphorylation. While this upstream motif does not mediate GRK recruitment, it appears to enhance GRK2 stability at MOR, suggesting that multisite phosphorylation is dependent on sustained GRK binding [16]. This indicates a potential role for GPCR phosphorylation sites in reinforcing GRK binding following initial recruitment mechanisms.

Recent work identified a high-energy electrostatic interaction, an “ionic lock”, between the GRK RH bundle and kinase C-lobe that maintains the kinase in an inactive conformation (Figure 3A). RH domain dissociation from the kinase domain (RH domain opening) is coupled to kinase domain closure and catalytic activation (Figure 3B and andC)C) [13,17]. GPCR-mediated disruption of the GRK5 ionic lock is associated with RH domain opening. Elongation of the kinase affords the RH bundle subdomain to further interact with the receptor, potentially stabilizing this active conformation [13]. Interestingly, disruption of the GRK1 ionic lock did not enhance rhodopsin recruitment [14]. This suggests that RH domain opening does not directly mediate GPCR binding but instead is likely a response to receptor interaction. Ultimately, these findings further illustrate how GPCRs promote GRK activation following initial recruitment.

The GRK αN-helix has been shown to be critical for GPCR phosphorylation [18] although its direct role in complex formation is unclear since it was not identified as a binding determinant in recent structural studies [13,14]. This suggests that it might be involved in GPCR-mediated kinase activation. Indeed, the ordered GRK6 N-terminus coordinates the kinase C-tail at the active site, a motif critical for catalysis [19]. In addition, a cytoplasmic hydrophobic pocket in rhodopsin is required for phosphorylation by GRK1 and modeling of the GRK1 αN-helix within this pocket suggests that the hydrophobic environment stabilizes the α-helix structure [20]. Thus, transient interfacing of the GRK N-terminus with the receptor cytoplasmic pocket during GRK recruitment may be a key step in the mechanism of GPCR-mediated kinase activation.

Regulation of GRKs

Various post-translational modifications including phosphorylation, ubiquitination and nitrosylation have been shown to regulate GRK activity, localization and degradation [2147] (Table 1). In addition, phospholipids mediate several aspects of GRK-mediated receptor phosphorylation and acidic lipids have been characterized as a key regulator, as reviewed previously [2]. PIP2 binding has been shown to orient GRKs at a model membrane plane, suggesting that PIP2 positions the kinase to effectively phosphorylate the receptor [48,49]. In fact, acidic lipids are necessary to form a stable GRK5/β2AR complex in vitro [13]. Additionally, Gβɣ mediates GRK2 subfamily phosphorylation of GPCRs, primarily by promoting membrane association via the PH domain [50]. The GRK2 subfamily RH domain also binds Gαq, sequestering G protein activity [51]. Recent evidence suggests that this Gαq interaction may also mediate GRK2 membrane binding to promote stable kinase association with the M3-muscarinic acetylcholine receptor (M3AChR) and subsequent arrestin recruitment [52].

Table 1:

GRK post-translational modifications that regulate kinase function

EnzymeGRKFunctional EffectMechanismSignalingMethodsReference
Cbl-c2DegradationUbiquitination via UCA1 bindingIncreased lncRNA UCA1NCI-N87, MGC-803 and HGC-27 cells[21]
CUL4-ROC1-DDB15DegradationUbiquitinationIrradiation/protein instabilityHEK293T cells[22]
2DegradationUbiquitination via Gβ bindingβAR, Smo inhibits Gβ/DDB1 complex via PKAin vitro, HEK293 and S2 cells, Human blood cells, Rat cardiomyocytes[2325]
MDM22DegradationUbiquitinationβAR via β-arrestin, IGFR via AktHEK293 cells, Mouse cardiac tissue[26,27]
NOS2Catalytic inhibitionNO-Cys340 (kinase domain)β2AR, Bradykinin receptor, P2YRin vitro, HEK293 cells, HUVEC, Mouse lung tissue[28]
PKA1Inhibition of GPCR phosphorylationpSer21 (N-terminus)Dark-dependent cAMPin vitro, HEK293 cells, Mouse retinal tissue[29,30]
7Inhibition of GPCR phosphorylationpSer23/Ser36 (N-terminus)Dark-dependent cAMPin vitro, HEK293 cells, x. laevis retinal tissue[29,31]
2Membrane recruitment via Gβɣ bindingpSer685 (C-terminus)β2AR, VPAC2HEK293 cells[3234]
PKC2Membrane recruitmentPhosphorylation within PH domain/C-terminusα1AR, ErGPCR-2 (H. armigera steroid hormone receptor)in vitro, HEK293, CHO and H. armigera epidermal cells[35,36]
2GPCR phosphorylationpSer29 (N-terminus)in vitro[37]
5Catalytic inhibition, inhibition of GPCR recruitmentPhosphorylation in C-terminusin vitro, COS-1 cells[38]
ERK2Catalytic inhibitionpSer670 (C-terminus)β2ARin vitro, HEK293 and COS-7 cells[39,40]
2Degradation via MDM2pSer670 (C-terminus)βAR via β-arrestinHEK293, COS-7 and MEF cells[41,42]
CDK22Degradation via Pin1 bindingpSer670 (C-terminus)G2/M cell cycle progressionin vitro, Hela, HEK293, MEF and HUVEC cells[43]
Src2Catalytic activationpTyr13/86/92 (N-terminus/RH domain)β2AR, TCR, EGFRin vitro, HEK293, COS-7 and Jurkat T cells[4446]
2DegradationpTyr13/86/92 (N-terminus/RH domain)β2AR, CXCR4 via β-arrestinHEK293, Jurkat T, C6, and COS-7 cells[42,47]

A recent study also established copper metabolism MURR1 domain–containing (COMMD) 3 and COMMD8 as adaptor proteins that mediate GRK6-specific recruitment to active GPCRs [53]. In this study, Nakai et al. identified GRK2/3-mediated phosphorylation of CXCR4 as a key determinant for recruitment of the COMMD3/8 complex. COMMD3/8 binding to the receptor subsequently recruits GRK6 and mediates GRK6-specific phosphorylation of CXCR4, initiating β-arrestin binding. Importantly, the authors demonstrate that this COMMD3/8-dependent CXCR4 signaling pathway mediates B-cell trafficking and the humoral immune response [53]. COMMD3/8 are ubiquitously expressed, therefore this complex may serve a general role for GRK6 recruitment to a range of GPCRs.

While GRKs are regulated by various post-translational modifications that can alter kinase stability and function (Table 1), several proteins have also been shown to directly inhibit GRK receptor phosphorylation. For example, raf kinase inhibitor protein (RKIP) is an established physiological inhibitor of GRK2-mediated GPCR phosphorylation and internalization, attenuating receptor desensitization. Phosphorylation of RKIP by protein kinase C (PKC) promotes RKIP binding to the GRK2 N-terminal region (residues 1–185). Thus, GPCR-mediated PKC activation tempers GRK2 receptor phosphorylation via RKIP and serves as a feedback mechanism to inhibit GPCR desensitization [54]. Recently, RKIP regulation of GRK2 was identified as the mechanism in which inflammatory signals sensitize the δ-opioid receptor (DOR). In peripheral neurons, GRK2 is constitutively bound to DOR, blocking G protein coupling. B2 bradykinin receptor stimulation via the inflammatory signal bradykinin activates PKC, switching RKIP binding to GRK2 and restoring DOR responsiveness [55]. RKIP regulation of GRK2 may also enhance GPCR signaling independent of receptor phosphorylation. For example, RKIP overexpression in mice induces heart failure progression that is partially dependent on angiotensin II receptor type 1 (AT1)-mediated Gq signaling. RKIP sequestration of the GRK2 RH domain is proposed to block GRK2 inhibition of Gq, restoring AT1-Gq coupling for cardiotoxic signaling [56]. Together, these recent studies establish that in addition to GPCR feedback signaling, RKIP regulation of GRK2 can mediate receptor crosstalk pathways as well as G protein coupling.

Several calcium binding proteins such as recoverin and calmodulin (CaM) were previously established as calcium-dependent inhibitors of GRK-mediated GPCR phosphorylation [57]. Recoverin binds GRK1 via the αN-helix and sequesters the kinase from phosphorylating rhodopsin [58]. CaM is selective for the GRK4 subfamily with the highest efficacy towards GRK5 although it can also inhibit GRK2. In addition, GRKs are thought to contain two distant and distinct CaM binding sites [57]. Despite CaM inhibition of GRK-mediated GPCR phosphorylation, CaM has been shown to stimulate GRK5 phosphorylation of soluble substrates [59]. Several structural and biophysical methods were recently employed to help define the structural basis of GRK5 regulation by CaM. While CaM was determined to form an equimolar complex with GRK5 [60], the precise structural mechanism in which CaM occupies two binding sites remains unclear. Interestingly, CaM also mediates non-canonical GRK5 activity implicated in cardiac hypertrophy [61], although its role in regulating GPCR signaling has not been determined. A recent study suggests that CaM binding inhibits GRK5 phosphorylation of MOR upon calcium channel influx [62]. Similar to RKIP, calcium-dependent CaM regulation of GRKs may serve to potentiate GPCR signaling when coupled to calcium activation, though additional exploration is necessary.

Role of GRKs in Arrestin Recruitment and GPCR Signaling

While arrestins play a key role in GPCR desensitization, they also mediate GPCR signaling via a range of arrestin-dependent pathways. Differential GPCR phosphorylation patterns by distinct GRK isoforms, or “barcoding”, drives the diversity of GPCR signaling by dictating arrestin conformation following recruitment. Barcoding by GRKs has been shown to modulate signaling across a range of GPCR substrates, as reviewed previously [63]. Conversely, it has been difficult to reconcile how a highly variable GPCR C-terminal phosphorylation sequence mediates a conserved mechanism of arrestin recruitment. The structural basis of how specific patterns of GPCR phosphorylation mediate arrestin recruitment and signaling has not been well characterized. Thus, two recent studies set out to define general GPCR phosphorylation codes and how they relate to arrestin affinity, activation, specificity and conformation using several structural and biochemical techniques [64,65]. Their efforts to generalize these phosphorylation patterns and their corresponding functions are critical towards understanding the complex nature of arrestin-dependent GPCR signaling. Ultimately, defining these barcodes will further inform how different GRKs regulate arrestin recruitment and diversify signaling.

Canonical GPCR signaling consists of balanced G protein activation, GRK-mediated phosphorylation and arrestin-dependent processes. In certain instances, either G protein or arrestin activation is dominant resulting in signaling that is biased towards a particular pathway. Prior studies have focused on circumstances that modulate G protein or arrestin recruitment as determinants for GPCR bias. Recently, GRKs have emerged as modulators of biased signaling due to their crucial role in arrestin recruitment through GPCR phosphorylation [66]. GPCR mutants that exhibit bias are useful models to tease apart G protein- or arrestin-specific pathways and are also valuable for understanding the mechanisms of biased signaling. For example, the G protein-biased β2AR mutant Y129A is unable to recruit arrestin. Further characterization of the receptor identified that the G protein-bias was due to a deficiency in phosphorylation by GRKs, ultimately inhibiting arrestin-dependent function [67]. Similarly, a G protein-biased dopamine D2 receptor (D2R) mutant deficient in arrestin recruitment also exhibited reduced GRK2 recruitment [68]. In addition, mutation of M3AChR phosphorylation sites elicits G protein bias [69]. This study identified receptor phosphorylation as a requirement for bronchial smooth muscle contraction, a response implicated in asthma. While GRKs were not specifically probed, the study demonstrates how a model receptor can inform the therapeutic outcomes of a potential ligand biased against receptor phosphorylation. Alternatively, an arrestin-biased D2R mutant unable to engage G proteins was dependent on GRK2 activity for receptor phosphorylation and arrestin recruitment [68]. Furthermore, GRK2 recruitment and function at an arrestin-biased D2R is G protein-independent, a component intrinsic to wildtype D2R balanced signaling. Corroborating results with a D2R arrestin-biased agonist (UNC9994) indicate that D2R may directly recruit GRK2 to mediate arrestin-biased signaling [68]. These studies suggest a larger, understudied contribution of GRKs to mechanisms of GPCR bias and underscore the necessity of GRK assessment when characterizing biased ligands.

GRK function can also dictate physiological GPCR bias due to its role in receptor signaling and trafficking pathways. It has been shown that protease activated receptor-1 (PAR1) activation by Activated Protein C results in arrestin-biased signaling that mediates a cytoprotective response in endothelial cells [70]. GRK5 is required for this cytoprotective pathway, suggesting that it mediates arrestin-biased signaling through PAR1 [71]. Regulation of GRK recruitment to the receptor can also modulate arrestin-mediated pathways, introducing receptor bias. For example, calcium influx via calcium channel activation inhibits the phosphorylation and internalization of MOR and is correlated with reduced GRK5 membrane localization. The authors suggest that Ca2+/CaM blocks receptor internalization by sequestering GRK5 to prevent MOR phosphorylation and subsequent arrestin recruitment [62]. Gastric inhibitory polypeptide receptor (GIPR) is a constitutively internalized and recycled receptor that mediates incretin signaling in adipocytes. As a result, the receptor undergoes a noncanonical desensitization pathway. Upon stimulation, GIPR is regulated via trafficking of the internalized endosomal receptor to the Golgi, ultimately slowing receptor recycling back to the plasma membrane. Analogous to the canonical desensitization mechanism, arrestin mediates GIPR targeting to the Golgi, effectively sequestering rapid receptor recycling. This pathway is dependent on both GRK2 and GRK5, likely via phosphorylation of multiple GIPR C-terminal sites. Interestingly, a GIPR variant that is associated with obesity shows enhanced sequestration upon stimulation. This variant exhibits increased arrestin binding that is independent of GRK receptor phosphorylation. Phosphorylation-independent binding is speculated to prolong arrestin at GIPR, ultimately biasing the receptor towards Golgi trafficking rather than immediate recycling [72]. Clearly, GRK activity serves a key role in initiating and moderating receptor regulatory pathways, biasing GPCR signaling.

Phosphorylation-independent regulation of GPCRs by GRKs is an emerging concept in receptor desensitization, primarily characterized by GRK2 sequestration of downstream effectors, such as Gαq or MEK [73]. Transient, phosphorylation-independent desensitization by GRK2 was observed for MOR, though via a distinct and novel mechanism [16]. MOR desensitization in the absence of GRK2/3 kinase activity was initially observed in neurons [74]. Additional mechanistic studies revealed that GRK2 and arrestin are both recruited to active MOR in the absence of receptor phosphorylation. This mode of arrestin recruitment is rapid and transient, initiating desensitization, but not endocytosis. Miess et al. suggest that GRK2 still regulates this rapid component of arrestin recruitment and desensitization, independent of kinase activity [16]. This function of GRK may immediately modulate MOR activity, serving as a short-term response until the receptor is phosphorylated by GRK2 for sustained regulation. In sensory neurons, GRK2 is basally associated with DOR, constitutively desensitizing the receptor by inhibiting G protein coupling in a kinase-independent manner. It is suggested that neither DOR phosphorylation nor arrestin regulate receptor signaling in this setting. Ultimately, inhibition of GRK2 binding to DOR sensitizes the receptor to stimulation [55]. Direct, phosphorylation-independent regulation of GPCRs by GRKs could constitute an alternate and more rapid form of desensitization that is also readily reversible. It is still unclear exactly how GRKs mediate this mechanism of GPCR regulation and whether it occurs outside the opioid receptor family, warranting further study.

GRK Inhibitors

Previous studies (reviewed in [75]) identified a number of GRK-specific inhibitors that have proven useful in dissecting GRK function and potentially biasing GPCR signaling. For example, GRK inhibition via Paroxetine or Cmpd101 attenuates βAR and MOR phosphorylation, blunting arrestin recruitment and receptor endocytosis in heterologous systems [74,76]. In addition, Cmpd101 effectively blocks MOR desensitization in rat neurons [74]. Paroxetine and Cmpd101 inhibition of GRK2 also prevent P2Y2 and histamine H1 receptor desensitization in rat and human smooth muscle cells, respectively. Ultimately, blocking GPCR desensitization via GRK2 inhibition maintains arterial contractions, suggesting that GRKs serve as a viable pharmacological target to mediate arterial tone and blood pressure [77].

Recent efforts in inhibitor development have focused on understanding the determinants for GRK potency and selectivity to improve on the therapeutic potential of existing inhibitors. Structural analysis revealed an active site hydrophobic pocket that can be targeted to enhance GRK2 inhibitor potency and selectivity [78]. It is thought that inhibitors with minimal hydrogen bonding at the active site hinge exhibit additional selectivity by affording structural flexibility. These compounds might avoid locking the kinase in the highly conserved active state and instead can access and stabilize a unique conformation. This strategy could circumvent the lack of structural diversity at the active site by instead targeting conformational diversity to achieve GRK and GRK subfamily selectivity [75,79]. Despite the success in identifying GRK2-selective compounds, it has been particularly difficult to identify a potent, GRK4 subfamily-specific inhibitor [80]. In one study, high-throughput screening and substituent modification yielded a GRK5 inhibitor with nanomolar potency, but with minimal selectivity over GRK2 [81]. Recently, Rowlands et al. utilized a unique GRK5 C-tail cysteine residue (Cys474) positioned at the active site to selectively target GRK5. The addition of an electrophilic moiety to an existing GRK inhibitor scaffold resulted in the covalent modification of Cys474. This electrophilic substituent is associated with enhanced GRK5 potency and specificity over GRK2. Therefore, irreversible GRK5 modification via Cys474 is a potentially effective method of gaining subfamily specificity in inhibitor development [82].

Conclusions

GRK function and regulation is fundamental to GPCR signaling. Over the past several years, structural detail has contributed to our understanding of the mechanisms of GPCR targeting and kinase activation. In addition, various forms of physiological and pharmacological GRK inhibition have been detailed, ultimately blocking GPCR desensitization. Several novel modes of GRK regulation of GPCR signaling were recently identified and GRK receptor phosphorylation was established as a determinant of receptor bias. Thus, continued insight into the mechanisms in which GRKs modulate GPCRs is critical to our understanding of receptor signaling.

Acknowledgements

This work was supported by NIH awards R35GM122541, R01HL142310, R01HL136219 and P01HL114471 (to J.L.B).

Footnotes

Declaration of interest

The authors declare no competing interests.

Declaration of interests: none

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

1. Gurevich VV, Gurevich EV: GPCR signaling regulation: the role of GRKs and arrestins. Front Pharmacol 2019, 10:125. [PMC free article] [PubMed] [Google Scholar]
2. Gurevich EV, Tesmer JJG, Mushegian A, Gurevich VV: G protein-coupled receptor kinases: more than just kinases and not only for GPCRs. Pharmacol Ther 2012, 133:40–69. [PMC free article] [PubMed] [Google Scholar]
3. Weller M, Virmaux N, Mandel P: Light stimulated phosphorylation of rhodopsin in the retina: the presence of a protein kinase that is specific for photobleached rhodopsin. Proc Natl Acad Sci U S A 1975, 72:381–385. [PMC free article] [PubMed] [Google Scholar]
4. Benovic JL, Strasser RH, Caron MG, Lefkowitz RJ: β-Adrenergic receptor kinase: Identification of a novel protein kinase that phosphorylates the agonist-occupied form of the receptor. Proc Natl Acad Sci U S A 1986, 83:2797–2801. [PMC free article] [PubMed] [Google Scholar]
5. Benovic JL, Deblasi A, Stone WC, Caron MG, Lefkowitz RJ: β-Adrenergic receptor kinase: primary structure delineates a multigene family. Science 1989, 246:235–240. [PubMed] [Google Scholar]
6. Benovic JL, Onorato JJ, Arriza JL, Stone WC, Lohse M, Jenkins NA, Gilbert DJ, Copeland NG, Caron MG, Lefkowitz RJ: Cloning, expression, and chromosomal localization of β-adrenergic receptor kinase 2. A new member of the receptor kinase family. J Biol Chem 1991, 266:14939–14946. [PubMed] [Google Scholar]
7. Lorenz W, Inglese J, Palczewski K, Onorato JJ, Caron MG, Lefkowitz RJ: The receptor kinase family: primary structure of rhodopsin kinase reveals similarities to the β-adrenergic receptor kinase. Proc Natl Acad Sci U S A 1991, 88:8715–8719. [PMC free article] [PubMed] [Google Scholar]
8. Ambrose C, James M, Barnes G, Lin C, Bates G, Altherr M, Duyao M, Groot N, Church D, Wasmuth JJ, et al.: A novel G protein-coupled receptor kinase gene cloned from 4p16.3. Hum Mol Genet 1992, 1:697–703. [PubMed] [Google Scholar]
9. Kunapuli P, Benovic JL: Cloning and expression of GRK5: a member of the G protein-coupled receptor kinase family. J Biol Chem 1993, 90:5599–5592. [PMC free article] [PubMed] [Google Scholar]
10. Benovic JL, Gomez J: Molecular cloning and expression of GRK6. a new member of the G protein-coupled receptor kinase family. J Biol Chem 1993, 268:19521–19527. [PubMed] [Google Scholar]
11. Hisatomi O, Matsuda S, Satoh T, Kotaka S, Imanishi Y, Tokunaga F: A novel subtype of G-protein-coupled receptor kinase, GRK7, in teleost cone photoreceptors. FEBS Lett 1998, 424:159–164. [PubMed] [Google Scholar]
12. Komolov KE, Benovic JL: G protein-coupled receptor kinases: past, present and future. Cell Signal 2018, 41:17–24. [PMC free article] [PubMed] [Google Scholar]
13. Komolov KE, Du Y, Duc NM, Betz RM, Rodrigues JPGLM, Leib RD, Patra D, Skiniotis G, Adams CM, Dror RO, et al.: Structural and functional analysis of a β2-adrenergic receptor complex with GRK5. Cell 2017, 169:407–421. [PMC free article] [PubMed] [Google Scholar]
**The authors present the first experimentally determined model of a GRK-GPCR complex, illustrating how GRKs engage with the active receptor. The study also characterizes a novel GRK domain interface and conformational change as components of the GPCR-mediated GRK activation mechanism. This work significantly advances our structural understanding of the GRK-GPCR interaction axis, a critical determinant of receptor function.
14. He Y, Gao X, Goswami D, Hou L, Pal K, Yin Y, Zhao G, Ernst OP, Griffin P, Melcher K, et al.: Molecular assembly of rhodopsin with G protein-coupled receptor kinases. Cell Res 2017, 27:728–747. [PMC free article] [PubMed] [Google Scholar]
15. Baameur F, Morgan DH, Yao H, Tran TM, Hammitt RA, Sabui S, Mcmurray JS, Lichtarge O, Clark RB: Role for the regulator of G-protein signaling homology domain of G protein-coupled receptor kinases 5 and 6 in β2-adrenergic receptor and rhodopsin phosphorylation. Mol Pharmacol 2010, 77:405–415. [PMC free article] [PubMed] [Google Scholar]
16. Miess E, Gondin AB, Yousuf A, Steinborn R, Mösslein N, Yang Y, Göldner M, Ruland JG, Bünemann M, Krasel C, et al.: Multisite phosphorylation is required for sustained interaction with GRKs and arrestins during rapid μ-opioid receptor desensitization. Sci Signal 2018, 11:eaas9609. [PubMed] [Google Scholar]
**This paper describes a novel mechanism of phosphorylation-independent μ-opioid receptor desensitization by GRK2 and how receptor phosphorylation patterns can dictate GRK receptor targeting. These findings indicate that GRK-mediated GPCR regulation may be more complex than currently appreciated.
17. Yao X, Cato MC, Labudde E, Beyett TS, Tesmer JJG: Navigating the conformational landscape of G protein-coupled receptor kinases during allosteric activation. J Biol Chem 2017, 292:16032–16043. [PMC free article] [PubMed] [Google Scholar]
18. Pao CS, Barker BL, Benovic JL: Role of the amino terminus of G protein-coupled receptor kinase 2 in receptor phosphorylation. Biochemistry 2009, 48:7325–7333. [PMC free article] [PubMed] [Google Scholar]
19. Boguth CA, Singh P, Huang C, Tesmer JJG: Molecular basis for activation of G protein-coupled receptor kinases. EMBO J 2010, 29:3249–3259. [PMC free article] [PubMed] [Google Scholar]
20. Jones Brunette AM, Sinha A, David L, Farrens DL: Evidence that the rhodopsin kinase (GRK1) N-terminus and the transducin Gα C-terminus interact with the same “hydrophobic patch” on rhodopsin TM5. Biochemistry 2016, 55:3123–3135. [PubMed] [Google Scholar]
21. Wang Z, He C, Hu L, Shi H, Li J, Gu Q, Su L, Liu B, Li C, Zhu Z: Long noncoding RNA UCA1 promotes tumour metastasis by inducing GRK2 degradation in gastric cancer. Cancer Lett 2017, 408:10–21. [PubMed] [Google Scholar]
22. Wu Z, Chen Y, Yang T, Gao Q, Yuan M, Ma L: Targeted ubiquitination and degradation of G-protein-coupled receptor kinase 5 by the DDB1-CUL4 ubiquitin ligase complex. PLoS One 2012, 7:e43997. [PMC free article] [PubMed] [Google Scholar]
23. Zha Z, Han X, Smith MD, Liu Y, Giguère PM, Kopanja D, Raychaudhuri P, Siderovski DP, Guan KL, Lei QY, et al.: A non-canonical function of Gβ as a subunit of E3 ligase in targeting GRK2 ubiquitylation. Mol Cell 2015, 58:794–803. [PMC free article] [PubMed] [Google Scholar]
24. Zha Z, Han X, Smith MD, Lei Q, Guan K, Xiong Y: Hypertension-associated C825T polymorphism impairs the function of Gβ3 to target GRK2 ubiquitination. Cell Discov 2016, 2:16005. [PMC free article] [PubMed] [Google Scholar]
25. Li S, Cho YS, Wang B, Li S, Jiang J: Regulation of Smoothened ubiquitylation and cell surface expression through a Cul4-DDB1-Gβ E3 ubiquitin ligase complex. J Cell Sci 2018, 131:jcs218016. [PMC free article] [PubMed] [Google Scholar]
26. Salcedo A, Mayor F, Penela P: Mdm2 is involved in the ubiquitination and degradation of G-protein-coupled receptor kinase 2. EMBO J 2006, 25:4752–4762. [PMC free article] [PubMed] [Google Scholar]
27. Jean-Charles PY, Yu SMW, Abraham D, Kommaddi RP, Mao L, Strachan RT, Zhang ZS, Bowles DE, Brian L, Stiber JA, et al.: Mdm2 regulates cardiac contractility by inhibiting GRK2-mediated desensitization of β-adrenergic receptor signaling. JCI insight 2017, 2:e95998. [PMC free article] [PubMed] [Google Scholar]
28. Whalen EJ, Foster MW, Matsumoto A, Ozawa K, Violin JD, Que LG, Nelson CD, Benhar M, Keys JR, Rockman HA, et al.: Regulation of β-adrenergic receptor signaling by S-nitrosylation of G-protein-coupled receptor kinase 2. Cell 2007, 129:511–522. [PubMed] [Google Scholar]
29. Horner TJ, Osawa S, Schaller MD, Weiss ER: Phosphorylation of GRK1 and GRK7 by cAMP-dependent protein kinase attenuates their enzymatic activities. J Biol Chem 2005, 280:28241–28250. [PubMed] [Google Scholar]
30. Kolesnikov AV, Chrispell JD, Osawa S, Kefalov VJ, Weiss ER: Phosphorylation at serine 21 in G protein-coupled receptor kinase 1 (GRK1) is required for normal kinetics of dark adaption in rod but not cone photoreceptors. FASEB J 2020, 34:2677–2690. [PMC free article] [PubMed] [Google Scholar]
31. Osawa S, Jo R, Weiss ER: Phosphorylation of GRK7 by PKA in cone photoreceptor cells is regulated by light. J Neurochem 2008, 107:1314–1324. [PMC free article] [PubMed] [Google Scholar]
32. Cong M, Perry SJ, Lin FT, Fraser ID, Hu LA, Chen W, Pitcher JA, Scott JD, Lefkowitz RJ: Regulation of membrane targeting of the G protein-coupled receptor kinase 2 by protein kinase A and its anchoring protein AKAP79. J Biol Chem 2001, 276:15192–15199. [PubMed] [Google Scholar]
33. Murthy KS, Mahavadi S, Huang J, Zhou H, Sriwai W: Phosphorylation of GRK2 by PKA augments GRK2-mediated phosphorylation, internalization, and desensitization of VPAC2 receptors in smooth muscle. Am J Physiol - Cell Physiol 2008, 294:477–487. [PubMed] [Google Scholar]
34. Brackley AD, Sarrami S, Gomez R, Guerrero KA, Jeske NA: Identification of a signaling cascade that maintains constitutive δ-opioid receptor incompetence in peripheral sensory neurons. J Biol Chem 2017, 292:8762–8772. [PMC free article] [PubMed] [Google Scholar]
35. Winstel R, Freund S, Krasel C, Hoppe E, Lohse MJ: Protein kinase cross-talk: membrane targeting of the receptor kinase by protein kinase C. Proc Natl Acad Sci U S A 1996, 93:2105–2109. [PMC free article] [PubMed] [Google Scholar]
36. Zhao WL, Wang D, Liu CY, Zhao XF: G-protein-coupled receptor kinase 2 terminates G-protein-coupled receptor function in steroid hormone 20-hydroxyecdysone signaling. Sci Rep 2016, 6:1–13. [PMC free article] [PubMed] [Google Scholar]
37. Krasel C, Dammeier S, Winstel R, Mischak H, Lohse MJ: Phosphorylation of GRK2 by protein kinase C abolishes its inhibition by calmodulin. J Biol Chem 2001, 276:1911–1915. [PubMed] [Google Scholar]
38. Pronin AN, Benovic JL: Regulation of the G protein-coupled receptor kinase GRK5 by protein kinase C. J Biol Chem 1997, 272:3806–3812. [PubMed] [Google Scholar]
39. Pitcher JA, Tesmer JJG, Freeman JLR, Capel WD, Stone WC, Lefkowitz RJ: Feedback inhibition of G protein-coupled receptor kinase 2 (GRK2) activity by extracellular signal-regulated kinases. J Biol Chem 1999, 274:34531–34534. [PubMed] [Google Scholar]
40. Elorza A, Sarnago S, Mayor F: Agonist-dependent modulation of G protein-coupled receptor kinase 2 by mitogen-activated protein kinases. Mol Pharmacol 2000, 57:778–783. [PubMed] [Google Scholar]
41. Elorza A, Penela P, Sarnago S, Mayor F: MAPK-dependent degradation of G protein-coupled receptor kinase 2. J Biol Chem 2003, 278:29164–29173. [PubMed] [Google Scholar]
42. Nogués L, Salcedo A, Mayor F, Penela P: Multiple scaffolding functions of β-arrestins in the degradation of G protein-coupled receptor kinase. J Biol Chem 2011, 286:1165–1173. [PMC free article] [PubMed] [Google Scholar]
43. Penela P, Rivas V, Salcedo A, Mayor F: G protein-coupled receptor kinase 2 (GRK2) modulation and cell cycle progression. Proc Natl Acad Sci U S A 2010, 107:1118–1123. [PMC free article] [PubMed] [Google Scholar]
44. Sarnago S, Elorza A, Mayor F: Agonist-dependent phosphorylation of the G protein-coupled receptor kinase 2 (GRK2) by Src tyrosine kinase. J Biol Chem 1999, 274:34411–34416. [PubMed] [Google Scholar]
45. Dinkel BA, Kremer KN, Rollins MR, Medlyn MJ, Hedin KE: GRK2 mediates TCR-induced transactivation of CXCR4 and TCR–CXCR4 complex formation that drives PI3K/PREX1 signaling and T cell cytokine secretion. J Biol Chem 2018, 293:14022–14039. [PMC free article] [PubMed] [Google Scholar]
46. Sun N, Zhang X, Guo S, Le HT, Zhang X, Kim KM: Molecular mechanisms involved in epidermal growth factor receptor-mediated inhibition of dopamine D3 receptor signaling. Biochim Biophys Acta - Mol Cell Res 2018, 1865:1187–1200. [PubMed] [Google Scholar]
47. Penela P, Elorza A, Sarnago S, Mayor F. J: β-arrestin- and c-Src-dependent degradation of G-protein-coupled receptor kinase 2. EMBO J 2001, 20:5129–5138. [PMC free article] [PubMed] [Google Scholar]
48. Yang P, Homan KT, Li Y, Cruz-Rodríguez O, Tesmer JJG, Chen Z: Effect of lipid composition on the membrane orientation of the G protein-coupled receptor kinase 2-Gβ1γ2 complex. Biochemistry 2016, 55:2841–2848. [PMC free article] [PubMed] [Google Scholar]
49. Yang P, Glukhova A, Tesmer JJG, Chen Z: Membrane orientation and binding determinants of G protein-coupled receptor kinase 5 as assessed by combined vibrational spectroscopic studies. PLoS One 2013, 8:e82072. [PMC free article] [PubMed] [Google Scholar]
50. Touhara K: Binding of multiple ligands to pleckstrin homology domain regulates membrane translocation and enzyme activity of β-adrenergic receptor kinase. FEBS Lett 1997, 417:243–248. [PubMed] [Google Scholar]
51. Day PW, Carman CV, Sterne-Marr R, Benovic JL, Wedegaertner PB: Differential interaction of GRK2 with members of the Gαq family. Biochemistry 2003, 42:9176–9184. [PubMed] [Google Scholar]
52. Wolters V, Krasel C, Brockmann J, Bünemann M: Influence of Gαq on the dynamics of M3-acetylcholine receptor-G-protein-coupled receptor kinase 2 interaction. Mol Pharmacol 2015, 87:9–17. [PubMed] [Google Scholar]
53. Nakai A, Fujimoto J, Miyata H, Stumm R, Narazaki M, Schulz S, Baba Y, Kumanogoh A, Suzuki K: The COMMD3/8 complex determines GRK6 specificity for chemoattractant receptors. J Exp Med 2019, 216:1630–1647. [PMC free article] [PubMed] [Google Scholar]
*This study identifies an adaptor protein complex as a novel mechanism of GRK6 recruitment to GPCRs, potentially dictating site-specific receptor phosphorylation.
54. Lorenz K, Lohse MJ, Quitterer U, Strasse V: Protein kinase C switches the Raf kinase inhibitor from Raf-1 to GRK-2. Nature 2003, 426:574–579. [PubMed] [Google Scholar]
55. Brackley AD, Gomez R, Akopian AN, Henry MA, Jeske NA: GRK2 constitutively governs peripheral delta opioid receptor activity. Cell Rep 2016, 16:2686–2698. [PMC free article] [PubMed] [Google Scholar]
56. Wolf S, Abd Alla J, Quitterer U: Sensitization of the angiotensin II AT1 receptor contributes to RKIP-induced symptoms of heart failure. Front Med 2019, 6:359. [PMC free article] [PubMed] [Google Scholar]
57. Sallese M, Iacovelli L, Cumashi A, Capobianco L, Cuomo L, De Blasi A: Regulation of G protein-coupled receptor kinase subtypes by calcium sensor proteins. Biochim Biophys Acta 2000, 1498:112–121. [PubMed] [Google Scholar]
58. Higgins MK, Oprian DD, Schertler GFX: Recoverin binds exclusively to an amphipathic peptide at the N terminus of rhodopsin kinase, inhibiting rhodopsin phosphorylation without affecting catalytic activity of the kinase. J Biol Chem 2006, 281:19426–19432. [PubMed] [Google Scholar]
59. Pronin AN, Morris AJ, Surguchov A, Benovic JL: Synucleins are a novel class of substrates for G protein-coupled receptor kinases. J Biol Chem 2000, 275:26515–26522. [PubMed] [Google Scholar]
60. Beyett TS, Fraley AE, Labudde E, Patra D, Coleman RC, Eguchi A, Glukhova A, Chen Q, Williams RM, Koch WJ, et al.: Perturbation of the interactions of calmodulin with GRK5 using a natural product chemical probe. Proc Natl Acad Sci U S A 2019, 116:15895–15900. [PMC free article] [PubMed] [Google Scholar]
61. Gold JI, Martini JS, Hullmann J, Gao E, Chuprun JK, Lee L, Tilley DG, Rabinowitz JE, Bossuyt J, Bers DM, et al.: Nuclear translocation of cardiac G protein-coupled receptor kinase 5 downstream of select Gq-activating hypertrophic ligands is a calmodulin-dependent process. PLoS One 2013, 8:e57324. [PMC free article] [PubMed] [Google Scholar]
62. Scherer PC, Zaccor NW, Neumann NM, Vasavda C, Barrow R, Ewald AJ, Rao F, Sumner CJ, Snyder SH: TRPV1 is a physiological regulator of μ-opioid receptors. PNAS 2017, 114:13561–13566. [PMC free article] [PubMed] [Google Scholar]
63. Jean-Charles P-Y, Kaur S, Shenoy SK: G Protein-Coupled Receptor Signaling Through β-Arrestin-Dependent Mechanisms. J Cardiovasc Pharmacol 2017, 70:142–158. [PMC free article] [PubMed] [Google Scholar]
64. Zhou XE, He Y, de Waal PW, Gao X, Kang Y, Van Eps N, Yin Y, Pal K, Goswami D, White TA, et al.: Identification of Phosphorylation Codes for Arrestin Recruitment by G Protein-Coupled Receptors. Cell 2017, 170:457–469. [PMC free article] [PubMed] [Google Scholar]
65. Mayer D, Damberger FF, Samarasimhareddy M, Feldmueller M, Vuckovic Z, Flock T, Bauer B, Mutt E, Zosel F, Allain FHT, et al.: Distinct G protein-coupled receptor phosphorylation motifs modulate arrestin affinity and activation and global conformation. Nat Commun 2019, 10:1261. [PMC free article] [PubMed] [Google Scholar]
66. Gurevich VV, Gurevich EV: Biased GPCR signaling: possible mechanisms and inherent limitations. Pharmacol Ther 2020, Epub:ahead of print. [PMC free article] [PubMed] [Google Scholar]
67. Choi M, Staus DP, Wingler LM, Ahn S, Pani B, Capel WD, Lefkowitz RJ: G protein–coupled receptor kinases (GRKs) orchestrate biased agonism at the β2-adrenergic receptor. Sci Signal 2018, 11:eaar7084. [PubMed] [Google Scholar]
**The authors describe a deficiency in GRK receptor phosphorylation as an additional determinant of G protein-biased GPCR signaling. This study establishes that the regulation of GRK function at the receptor serves as a potential mechanism for biasing signaling.
68. Pack TF, Orlen MI, Ray C, Peterson SM, Caron MG: The dopamine D2 receptor can directly recruit and activate GRK2 without G protein activation. J Biol Chem 2018, 293:6161–6171. [PMC free article] [PubMed] [Google Scholar]
*The authors describe a potential form of arrestin-biased signaling that is characterized by G protein-independent recruitment of GRK2. This mechanism of bias could have implications for biased ligand development.
69. Bradley SJ, Wiegman CH, Iglesias MM, Kong KC, Butcher AJ, Plouffe B, Goupil E, Bourgognon JM, Macedo-Hatch T, Legouill C, et al.: Mapping physiological G protein-coupled receptor signaling pathways reveals a role for receptor phosphorylation in airway contraction. Proc Natl Acad Sci U S A 2016, 113:4524–4529. [PMC free article] [PubMed] [Google Scholar]
70. Soh UJK, Trejo JA: Activated protein C promotes protease-activated receptor-1 cytoprotective signaling through β-arrestin and dishevelled-2 scaffolds. Proc Natl Acad Sci U S A 2011, 108:E1372–E1380. [PMC free article] [PubMed] [Google Scholar]
71. Roy RV, Ardeshirylajimi A, Dinarvand P, Yang L, Rezaie AR: Occupancy of human EPCR by protein C induces β-arrestin-2 biased PAR1 signaling by both APC and thrombin. Blood 2016, 128:1884–1893. [PMC free article] [PubMed] [Google Scholar]
72. Abdullah N, Beg M, Soares D, Dittman JS, McGraw TE: Downregulation of a GPCR by β-Arrestin2-mediated switch from an endosomal to a TGN recycling pathway. Cell Rep 2016, 17:2966–2978. [PMC free article] [PubMed] [Google Scholar]
73. Evron T, Daigle TL, Caron MG: GRK2: multiple roles beyond G protein-coupled receptor desensitization. Trends Pharmacol Sci 2012, 33:154–164. [PMC free article] [PubMed] [Google Scholar]
74. Lowe JD, Sanderson HS, Cooke AE, Ostovar M, Tsisanova E, Withey SL, Chavkin C, Husbands SM, Kelly E, Henderson G, et al.: Role of G protein-coupled receptor kinases 2 and 3 in μ-opioid receptor desensitization and internalization. Mol Pharmacol 2015, 88:347–356. [PMC free article] [PubMed] [Google Scholar]
75. Homan KT, Tesmer JJG: Molecular basis for small molecule inhibition of G protein-coupled receptor kinases. ACS Chem Biol 2015, 10:246–256. [PMC free article] [PubMed] [Google Scholar]
76. Guo S, Carter RL, Grisanti LA, Koch WJ, Tilley DG: Impact of paroxetine on proximal β-adrenergic receptor signaling. Cell Signal 2017, 38:127–133. [PMC free article] [PubMed] [Google Scholar]
77. Rainbow RD, Brennan S, Jackson R, Beech AJ, Bengreed A, Waldschmidt HV, Tesmer JJG, Challiss RAJ, Willets JM: Small-molecule G protein-coupled receptor kinase inhibitors attenuate G protein-coupled receptor kinase 2-mediated desensitization of vasoconstrictor-induced arterial contractions. Mol Pharmacol 2018, 94:1079–1091. [PMC free article] [PubMed] [Google Scholar]
78. Waldschmidt HV, Homan KT, Cato MC, Cruz-Rodríguez O, Cannavo A, Wilson MW, Song J, Cheung JY, Koch WJ, Tesmer JJG, et al.: Structure-based design of highly selective and potent G protein-coupled receptor kinase 2 inhibitors based on paroxetine. J Med Chem 2017, 60:3052–3069. [PMC free article] [PubMed] [Google Scholar]
79. Bouley R, Waldschmidt HV, Cato MC, Cannavo A, Song J, Cheung JY, Yao XQ, Koch WJ, Larsen SD, Tesmer JJG: Structural determinants influencing the potency and selectivity of indazole-paroxetine hybrid G protein-coupled receptor kinase 2 inhibitors. Mol Pharmacol 2017, 92:707–717. [PMC free article] [PubMed] [Google Scholar]
80. Waldschmidt HV, Bouley R, Kirchhoff PD, Lee P, Tesmer JJG, Larsen SD: Utilizing a structure-based docking approach to develop potent G protein-coupled receptor kinase (GRK) 2 and 5 inhibitors. Bioorganic Med Chem Lett 2018, 28:1507–1515. [PMC free article] [PubMed] [Google Scholar]
81. Cho SY, Lee BH, Jung H, Yun CS, Ha J Du, Kim HR, Chae CH, Lee JH, Seo HW, Oh KS: Design and synthesis of novel 3-(benzo[d]oxazol-2-yl)-5-(1-(piperidin-4-yl) −1H-pyrazol-4-yl)pyridin-2-amine derivatives as selective G-protein-coupled receptor kinase-2 and −5 inhibitors. Bioorganic Med Chem Lett 2013, 23:6711–6716. [PubMed] [Google Scholar]
82. Rowlands RA, Cato MC, Waldschmidt HV, Bouley RA, Chen Q, Avramova L, Larsen SD, Tesmer JJG, White AD: Structure-based design of selective, covalent G protein-coupled receptor kinase 5 inhibitors. ACS Med Chem Lett 2019, 10:1628–1634. [PMC free article] [PubMed] [Google Scholar]
*This paper illustrates an alternative method of GRK5 inhibitor development that can enhance compound selectivity. This strategy circumvents the highly conserved GRK active site architecture that has complicated the identification of a selective GRK5 inhibitor.
-