G protein-coupled receptor kinases: More than just kinases and not only for GPCRs

https://doi.org/10.1016/j.pharmthera.2011.08.001Get rights and content

Abstract

G protein-coupled receptor (GPCR) kinases (GRKs) are best known for their role in homologous desensitization of GPCRs. GRKs phosphorylate activated receptors and promote high affinity binding of arrestins, which precludes G protein coupling. GRKs have a multidomain structure, with the kinase domain inserted into a loop of a regulator of G protein signaling homology domain. Unlike many other kinases, GRKs do not need to be phosphorylated in their activation loop to achieve an activated state. Instead, they are directly activated by docking with active GPCRs. In this manner they are able to selectively phosphorylate Ser/Thr residues on only the activated form of the receptor, unlike related kinases such as protein kinase A. GRKs also phosphorylate a variety of non-GPCR substrates and regulate several signaling pathways via direct interactions with other proteins in a phosphorylation-independent manner. Multiple GRK subtypes are present in virtually every animal cell, with the highest expression levels found in neurons, with their extensive and complex signal regulation. Insufficient or excessive GRK activity was implicated in a variety of human disorders, ranging from heart failure to depression to Parkinson's disease. As key regulators of GPCR-dependent and -independent signaling pathways, GRKs are emerging drug targets and promising molecular tools for therapy. Targeted modulation of expression and/or of activity of several GRK isoforms for therapeutic purposes was recently validated in cardiac disorders and Parkinson's disease.

Introduction

Signaling via G protein-coupled receptors (GPCR) is terminated by a remarkably uniform two-step mechanism: a GPCR kinase (GRK) phosphorylates the active receptor, converting it into a target for high affinity binding of arrestin. Bound arrestin shields the cytoplasmic surface of the receptor, precluding G protein binding and activation (Wilden, 1995, Krupnick et al., 1997).

Phosphorylation of rhodopsin, a prototypical GPCR, upon its activation by light was first described in 1972 (Bownds et al., 1972, Kühn and Dreyer, 1972). Soon thereafter “opsin kinase” (modern name GRK11), which selectively phosphorylates active rhodopsin, was identified (Weller et al., 1975). The first clear evidence that rhodopsin phosphorylation is necessary for its rapid deactivation was presented in 1980 and led to the hypothesis that this mechanism may also regulate hormone receptors (Liebman & Pugh, 1980). Within a few years, this idea was confirmed for β2-adrenergic receptor (β2AR) (Stadel et al., 1983, Sibley et al., 1985) and later for many others (reviewed in Carman & Benovic, 1998). The demonstration of sequence similarity between the β2AR and rhodopsin in 1986 (Dixon et al., 1986) led to the recognition of the family of G protein-coupled receptors (GPCRs), of which rhodopsin is a founding member. Also in 1986, a kinase that could phosphorylate activated β-adrenergic receptors (βARK; modern name GRK2) was identified (Benovic et al., 1986b). This enzyme could also phosphorylate rhodopsin in a light-dependent manner (Benovic et al., 1986a). Phosphorylation of rhodopsin facilitates the binding of another protein termed arrestin (called 48-kDa protein at the time), which physically blocks further signaling by the receptor to heterotrimeric G proteins (Wilden et al., 1986). Demonstration that desensitization of the β2AR requires a homolog of arrestin (Benovic et al., 1987) firmly established the paradigm of two-step GPCR inactivation, which was later shown to apply to the majority of GPCRs (Carman and Benovic, 1998, Gurevich and Gurevich, 2004, Gurevich and Gurevich, 2006b). The cloning of GRK2 in 1989 suggested that it belongs to a distinct lineage of eukaryotic Ser/Thr protein kinases (Benovic et al., 1989a) that are a subclass of the AGC kinase group (Manning et al., 2002). In rapid succession, the members of this family expanded to include βARK2 (GRK3) (Benovic et al., 1991), GRK4 (Ambrose et al., 1992), GRK5 (Kunapuli & Benovic, 1993), and GRK6 (Benovic & Gomez, 1993). Cone specific GRK7 (Hisatomi et al., 1998, Weiss et al., 1998) completed the set of vertebrate GRKs.

The expression of mammalian GRK1 and GRK7 is largely limited to vertebrate rod and cone photoreceptors although both are also present in pinealocytes (Somers and Klein, 1984, Zhao et al., 1997, Zhao et al., 1999, Pugh and Lamb, 2000). Virtually every mammalian cell expresses several isoforms of non-visual GRKs from early embryonic development. GRK4 is expressed at high levels only in testis (Premont et al., 1996). In addition, GRK4 expression was detected in proximal tubule cells in kidneys, where GRK4α and GRK4γ variants reportedly regulate the signaling of D1 and D3 dopamine receptors (Felder et al., 2002, Villar et al., 2009). GRK4 is also expressed in the brain (Sallese et al., 2000b) and uterus myometrium (Brenninkmeijer et al., 1999). In the rat brain, four GRK isoforms, GRKs 2, 3, 5, and 6, are found as early as embryonic day 14 (Gurevich et al., 2004). Unfortunately, the information about the cell-specific expression of GRK isoforms is limited. We mostly know their distribution at the tissue level. The cellular complement of GRK isoforms may prove to be the most important determinant of specificity in GRK function. For example, both GRK1 and GRK2 efficiently phosphorylate light-activated rhodopsin, but GRK2 does not perform this function in GRK1 knockout mice.

The importance of the GRK-mediated signal shutoff is best illustrated in the visual system, where the lack of GRK1 or sites for GRK phosphorylation on rhodopsin leads to the loss of photoresponses, photoreceptor degeneration, and blindness in mice and night blindness in humans (Chen, Makino, et al., 1995, Yamamoto et al., 1997, Khani et al., 1998, Chen et al., 1999, Zhang et al., 2005, Hayashi et al., 2007, Song et al., 2009, Fan et al., 2010). In other cell types, the results are not as dramatic, except in development. In Drosophila, Gprk2,2 an ortholog of GRK4/5/6, is required for wing morphogenesis (Molnar et al., 2007), egg morphogenesis, and embryogenesis (Schneider & Spradling, 1997). Knockdown of Grk2 in zebrafish embryos induces early developmental arrest (Jiang et al., 2009), and knockout of GRK2 in mice is embryonic lethal due to abnormal formation of the heart (Jaber et al., 1996). This lethality stems from general, albeit undefined, role of GRK2 in embryogenesis, rather than specific role in the heart development, because mice with GRK2 ablation specific to the cardiac myocytes develop normally (Matkovich et al., 2006).

We know about structure and function of GRKs a lot less than these proteins deserve, considering that GRKs critically influence the function of most GPCRs, which are the targets of a large percentage of clinically used drugs (Gruber et al., 2010). Many issues are far from resolved. GRK specificity towards particular receptor subtypes is one important unanswered question. As mammals have only five non-visual GRKs and >800 GPCRs (Gruber et al., 2010), there are hundreds of GPCRs per GRK. It follows that each GRK must have the ability to phosphorylate many different receptors. However, neither the level of receptor specificity nor actual preference for particular GPCRs of non-visual GRKs is clear. We also need to fully describe how active GPCRs activate GRKs, which would be greatly facilitated by a structure of a receptor–GRK complex. This would define the full receptor footprint on the GRK and provide greater insight into the mechanism of kinase activation. GRKs phosphorylate many non-GPCR substrates, but it remains unknown whether proteins other than GPCRs can activate GRKs.

GRKs 2 and 5 have long been considered promising therapeutic targets for cardiac diseases (Penela et al., 2006). However, there has been little research regarding their value as therapeutic targets for other conditions. We believe that GRKs, by virtue of their regulatory nature, hold a great promise for therapy of disorders involving an imbalance in GPCR signaling. However, a better understanding of their structure and function is a prerequisite for successful therapeutic intervention.

Section snippets

Structural organization of G protein-coupled receptor kinases

Based on sequence similarity and gene structure, vertebrate GRKs are classified into three subfamilies: GRK1 comprising GRK1 (rhodopsin kinase) and GRK7 (cone kinase), GRK2 comprising GRK2 and 3, and GRK4 comprising GRK4, 5, and 6 (Premont et al., 1999). All GRKs are multi-domain proteins (Fig. 1) consisting of ~25-residue N-terminal region unique to the GRK family of kinases, followed by the regulator of G protein signaling (RGS) homology domain (RH) (Siderovski et al., 1996), and a Ser/Thr

Subcellular targeting of G protein-coupled kinase isoforms

The various GRK subfamilies employ several distinct mechanisms that bring them to or retain them at the membrane, where their integral membrane substrates GPCRs are found (Fig. 1). Visual subtypes have characteristic CaaX motif on the C-terminus for prenylation: GRK1 is farnesylated (Inglese et al., 1992a), whereas GRK7 is geranylgeranylated (Hisatomi et al., 1998). The association of GRKs 1 and 7 with the membrane is mediated by C-terminal prenylation. Therefore, visual GRKs “search” for

Mechanism of activation of G protein-coupled receptor kinases by active G protein-coupled receptors

The ability to phosphorylate active GPCRs was the first GRK function to be discovered. Receptor phosphorylation by itself can decrease G protein coupling (Wilden, 1995) and enables high-affinity binding of arrestin, which stops G protein-mediated signaling by blocking the cytoplasmic surface of the receptors (Krupnick et al., 1997). The most striking feature distinguishing GRKs from other kinases is that their activity depends on the functional state of the target: GRKs effectively

G protein-coupled receptor kinases phosphorylate non-G protein-coupled receptor substrates

An intriguing development in recent years has been a discovery of the ability of GRKs to interact with a variety of proteins other than GPCRs and in many cases to phosphorylate them (Table 1). The data extend the repertoire of pathways whose signaling is controlled by GRKs via phosphorylation of various signaling components. The list of non-GPCR substrates now includes single transmembrane domain tyrosine kinases (PDGFRβ), single transmembrane domain serine/threonine kinases, death receptors,

Proteins regulated by G protein-coupled receptor kinases in phosphorylation-independent manner

GRKs have been reported to regulate several signaling proteins via direct interaction that does not require kinase activity (Table 2). It is not unusual for enzymes to perform scaffolding functions in addition to or instead of their enzymatic activity. GRKs, particularly GRK2 and 3, are fairly large multidomain proteins, and it is conceivable that they could interact with a multitude of proteins via different domains (Pronin et al., 1997, Lodowski et al., 2003, Tesmer et al., 2005). As the

Regulation of G protein-coupled receptor kinases

The expression level, as well as activity of most enzymes in the cell is tightly regulated. GRKs are no exception. As described above, the best-known mechanism of GRK regulation is via direct binding to active GPCRs. However, this is just one of several established regulatory mechanisms.

G protein-coupled kinase isoforms — more of the same?

No review of GRKs would be complete without discussion of receptor specificity of GRK isoforms. The situation with GRKs resembles that with arrestins (Gurevich & Gurevich, 2006a) in that there are too few of them to be strictly receptor specific. Thus, it is often assumed that GRK isoforms are “nonspecific” towards GPCRs. However, the issue is not as simple as it appears. There are just too many GRK isoforms preserved over millions of years of vertebrate and mammalian evolution to believe that

Physiological and pathological roles of G protein-coupled kinase isoforms

In spite of the obvious importance of GRKs for the regulation of the GPCR signaling, their physiological functions remain poorly understood. Although the vertebrate GRK family is not large – only 7 members – it is functionally quite diverse, with two members, GRKs 1 and 7, playing specialized roles in photoreceptor cells and others ubiquitously expressed throughout the body obviously fulfilling multiple needs. To sort out all these complex interlinked functions played by individual GRK isoforms

Conclusions and future prospects

GRKs are well-established regulators of signaling and trafficking of GPCRs, the most numerous class of cell surface receptors targeted by about half of clinically used drugs (Jacoby et al., 2006). Recent findings show that GRKs also regulate a number of other GPCR-independent signaling pathways intimately involved in many vital cellular functions. Despite their obvious importance, GRKs are under-appreciated as drug targets and therapeutic tools.

In order to take full advantage of GRKs as

Conflict of interest statement

The authors declare that there are no conflicts of interests.

Acknowledgment

This work was supported by NIH grants NS065868 (EVG), HL071818 (JJGT), EY011500, GM077561, GM081756 (VVG), and by Stowers Institute for Medical Research (ARM).

References (450)

  • B.M. Binder et al.

    Light activation of one rhodopsin molecule causes the phosphorylation of hundreds of others. A reaction observed in electropermeabilized frog rod outer segments exposed to dim illumination

    J Biol Chem

    (1990)
  • B.M. Binder et al.

    Phosphorylation of non-bleached rhodopsin in intact retinas and living frogs

    J Biol Chem

    (1996)
  • P. Bisegna et al.

    Diffusion of the second messengers in the cytoplasm acts as a variability suppressor of the single photon response in vertebrate phototransduction

    Biophys J

    (2008)
  • M.R. Bruchas et al.

    Kappa opioid receptor activation of p38 MAPK is GRK3- and arrestin-dependent in neurons and astrocytes

    J Biol Chem

    (2006)
  • E.R. Bychkov et al.

    Arrestins and two receptor kinases are upregulated in Parkinson's disease with dementia

    Neurobiol Aging

    (2008)
  • E. Bychkov et al.

    Reduced expression of G protein-coupled receptor kinases in schizophrenia but not in schizoaffective disorder

    Neurobiol Dis

    (2011)
  • C.V. Carman et al.

    G-protein-coupled receptors: turn-ons and turn-offs

    Curr Opin Neurobiol

    (1998)
  • C.V. Carman et al.

    Selective regulation of Galpha(q/11) by an RGS domain in the G protein-coupled receptor kinase, GRK2

    J Biol Chem

    (1999)
  • C.V. Carman et al.

    Binding and phosphorylation of tubulin by G protein-coupled receptor kinases

    J Biol Chem

    (1998)
  • L.A. Catapano et al.

    G protein-coupled receptors in major psychiatric disorders

    Biochim Biophys Acta

    (2007)
  • E.P. Chen et al.

    Myocardial function in hearts with transgenic overexpression of the G protein-coupled receptor kinase 5

    Ann Thorac Surg

    (2001)
  • C.Y. Chen et al.

    Beta-adrenergic receptor kinase. Agonist-dependent receptor binding promotes kinase activation

    J Biol Chem

    (1993)
  • C.-K. Chen et al.

    Ca2 + −dependent interaction of recoverin with rhodopsin kinase

    J Biol Chem

    (1995)
  • M. Chen et al.

    G Protein-coupled receptor kinases phosphorylate LRP6 in the Wnt pathway

    J Biol Chem

    (2009)
  • S. Cheng et al.

    Regulation of smoothened by Drosophila G-protein-coupled receptor kinases

    Dev Biol

    (2010)
  • T.T. Chuang et al.

    Phosphorylation and activation of b-adrenergic receptor kinase by protein kinase C

    J Biol Chem

    (1995)
  • T.T. Chuang et al.

    Inhibition of G protein-coupled receptor kinase subtypes by Ca2+/calmodulin

    J Biol Chem

    (1996)
  • P.M. Cobelens et al.

    Hydrogen peroxide impairs GRK2 translation via a calpain-dependent and cdk1-mediated pathway

    Cell Signal

    (2007)
  • M. Cong et al.

    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)
  • M.S. Cortese et al.

    Intrinsic disorder in scaffold proteins: getting more from less

    Prog Biophys Mol Biol

    (2008)
  • P.W. Day et al.

    Analysis of G-protein-coupled receptor kinase RGS homology domains

    Methods Enzymol

    (2004)
  • S.K. DebBurman et al.

    G protein-coupled receptor kinase GRK2 is a phospholipid-dependent enzyme that can be conditionally activated by G protein betagamma subunits

    J Biol Chem

    (1996)
  • S.K. DebBurman et al.

    Lipid-mediated regulation of G protein-coupled receptor kinases 2 and 3

    J Biol Chem

    (1995)
  • G.K. Dhami et al.

    Phosphorylation-independent regulation of metabotropic glutamate receptor signaling by G protein-coupled receptor kinase 2

    J Biol Chem

    (2002)
  • G.K. Dhami et al.

    G Protein-coupled receptor kinase 2 regulator of G protein signaling homology domain binds to both metabotropic glutamate receptor 1a and Galphaq to attenuate signaling

    J Biol Chem

    (2004)
  • A. Diaz et al.

    Regulation of mu-opioid receptors, G-protein-coupled receptor kinases and beta-arrestin 2 in the rat brain after chronic opioid receptor antagonism

    Neuroscience

    (2002)
  • M.R. Ahmed et al.

    Lentiviral overexpression of GRK6 alleviates L-dopa-induced dyskinesia in experimental Parkinson's disease

    Sci Transl Med

    (2010)
  • M.R. Ahmed et al.

    Altered expression and subcellular distribution of GRK subtypes in the dopamine-depleted rat basal ganglia is not normalized by l-DOPA treatment

    J Neurochem

    (2007)
  • M.R. Ahmed et al.

    Haloperidol and clozapine differentially affect the expression of arrestins, receptor kinases, and ERK activation

    J Pharmacol Exp Ther

    (2008)
  • C. Ambrose et al.

    A novel G protein-coupled receptor kinase gene cloned from 4p16.3

    Hum Mol Genet

    (1992)
  • J.B. Ames et al.

    Structural basis for calcium-induced inhibition of rhodopsin kinase by recoverin

    J Biol Chem

    (2006)
  • S. Andréasson et al.

    A six-generation family with autosomal dominant retinitis pigmentosa and a rhodopsin gene mutation (arginine-135-leucine)

    Ophthalmic Paediatr Genet

    (1992)
  • A.M. Aragay et al.

    Monocyte chemoattractant protein-1-induced CCR2B receptor desensitization mediated by the G protein-coupled receptor kinase 2

    Proc Natl Acad Sci U S A

    (1998)
  • I. Aramori et al.

    Molecular mechanism of desensitization of the chemokine receptor CCR-5: receptor signaling and internalization are dissociable from its role as an HIV-1 co-receptor

    EMBO J

    (1997)
  • S. Arawaka et al.

    The role of G-protein-coupled receptor kinase 5 in pathogenesis of sporadic Parkinson's disease

    J Neurosci

    (2006)
  • D. Arinobu et al.

    Larger inhibition of visual pigment kinase in cones than in rods

    J Neurochem

    (2010)
  • I. Aubert et al.

    Increased D1 dopamine receptor signaling in levodopa-induced dyskinesia

    Ann Neurol

    (2005)
  • F. Baameur et al.

    Role for the regulator of G-protein signaling homology domain of G protein-coupled receptor kinases 5 and 6 in beta 2-adrenergic receptor and rhodopsin phosphorylation

    Mol Pharmacol

    (2010)
  • A.A. Banday et al.

    Insulin causes renal dopamine D1 receptor desensitization via GRK2-mediated receptor phosphorylation involving phosphatidylinositol 3-kinase and protein kinase C

    Am J Physiol

    (2007)
  • T.B. Barrett et al.

    Further evidence for association of GRK3 to bipolar disorder suggests a second disease mutation

    Psychiatr Genet

    (2007)
  • Cited by (0)

    View full text